1
|
Vidal-Manrique M, Nieuwenstein T, Hooijmaijers L, de Jonge P, Djojoatmo M, Jansen J, van der Waart A, Brock R, Dolstra H. IL-15 transpresentation by ovarian cancer cells improves CD34 + progenitor-derived NK cell's anti-tumor functionality. Oncoimmunology 2025; 14:2465010. [PMID: 39960378 PMCID: PMC11834524 DOI: 10.1080/2162402x.2025.2465010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy. As high numbers of Natural Killer (NK) cells in ascites associate with improved survival, the adoptive transfer of allogeneic NK cells is an attractive therapeutic strategy. An approach to further improve NK cell expansion and anti-tumor functionality post-infusion includes IL-15 transpresentation (transIL-15), which involves surface expression of the IL-15 cytokine bound to IL-15Rα. However, others have substantiated that systemic administration of ALT/N-803, a soluble molecule mimicking transIL-15, leads to T cell-mediated rejection of the infused allogeneic NK cell product. In addition, whether transIL-15 induce superior expansion and functionality of our hematopoietic progenitor cell-derived NK cells (HPC-NK) remains understudied. Here, we propose to transfect OC cells with IL-15 and IL-15Rα mRNA and evaluate HPC-NK cell stimulation in vitro. Co-transfection of both mRNAs resulted in surface co-expression of both components, thus mimicking the transIL-15. Importantly, co-culture of HPC-NK cells with transIL-15 OC cells resulted in superior proliferation, IFNγ production, cytotoxicity and granzyme B secretion. Furthermore, we observed uptake of IL-15Rα by HPC-NK cells when co-cultured with transIL-15 OC cells, which associates with NK cell long-term proliferation and survival. Superior killing and granzyme B secretion were also observed in transIL-15 OC spheroids. Our results demonstrate that local delivery of IL-15 and IL-15Rα mRNA to OC tumors may be a safer strategy to boost HPC-NK cell therapy of OC through IL-15 transpresentation.
Collapse
Affiliation(s)
- M. Vidal-Manrique
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - T. Nieuwenstein
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L. Hooijmaijers
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - P.K.J.D. de Jonge
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M. Djojoatmo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J. Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A.B. van der Waart
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - R. Brock
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - H. Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Gudoityte G, Sharma O, Leuenberger L, Wallin E, Fernebro J, Östling P, Bergström R, Lindberg J, Joneborg U, Kallioniemi O, Seashore-Ludlow B. Systematic profiling of cancer-fibroblast interactions reveals drug combinations in ovarian cancer. Mol Oncol 2025. [PMID: 40411295 DOI: 10.1002/1878-0261.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/07/2025] [Accepted: 05/05/2025] [Indexed: 05/26/2025] Open
Abstract
Ovarian cancer (OC) is a leading cause of death of gynecological cancers in women. Poor patient response to treatment highlights the need to better understand how the tumor microenvironment affects OC progression. Growing evidence indicates the crucial role of non-cancerous components, such as cancer-associated fibroblasts, in establishing a complex network of cellular and molecular interactions, influencing cancer progression and response to treatment. Therefore, in this study, we sought to characterize the impact of fibroblasts on OC cell behavior and drug response. Using both direct and indirect cell co-culture systems, we observed distinct changes in cancer cell proliferation, morphology, and secretome in the presence of fibroblasts. Furthermore, an imaging-based high-throughput drug screen of 528 oncology compounds revealed multiple drugs that showed altered efficacy in the co-culture conditions, demonstrating the role of fibroblasts in driving cancer cell resistance to treatment. Most importantly, our data identified the two drug combinations of Birinapant or Vorinostat with Carboplatin as promising treatments, exploiting the altered cancer cell phenotype in co-cultures. These findings were supported by the increased sensitivity of ex vivo cultures to these combinations.
Collapse
Affiliation(s)
- Greta Gudoityte
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Osheen Sharma
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Laura Leuenberger
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Emelie Wallin
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pelvic Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Josefin Fernebro
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pelvic Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Päivi Östling
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Rebecka Bergström
- Department of Molecular Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Lindberg
- Department of Molecular Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Joneborg
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Olli Kallioniemi
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Institute for Molecular Medicine Finland, University of Helsinki, Finland
| | - Brinton Seashore-Ludlow
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Li H, Wang Y, Zhou S, Liu J, Jin Y. Apatinib-loaded silicate nanoparticles coated with macrophage membranes and PD-1 antibody for enhanced chemo-immunotherapy in ovarian cancer via VEGFR2 and PD-1 dual inhibition. Colloids Surf B Biointerfaces 2025; 254:114790. [PMID: 40381292 DOI: 10.1016/j.colsurfb.2025.114790] [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: 03/04/2025] [Revised: 04/25/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
Ovarian cancer remains one of the most challenging malignancies to treat due to its aggressive nature and resistance to conventional therapies. In this study, we developed a nanoparticle-based system (Apa@SiO2@MP) that combines chemotherapy with immune checkpoint inhibition for enhanced treatment of ovarian cancer. The system consists of mesoporous silica nanoparticles (SiO2 NPs) coated with macrophage membranes (MP) and functionalized with programmed death 1 (PD-1) antibody, designed to improve the delivery and targeting of apatinib, a tyrosine kinase inhibitor. The system demonstrated effective drug encapsulation, controlled release, and stability in physiological environments. In vitro assays revealed that Apa@SiO2@MP had minimal cytotoxicity in normal cells but significantly reduced cell viability in ovarian cancer cells (SKOV-3), highlighting its cancer-targeting ability. Apatinib effectively inhibited VEGFR2 expression and induced reactive oxygen species (ROS) production, further promoting anti-cancer effects. In vivo, Apa@SiO2@MP treatment led to enhanced tumor inhibition, as well as significant immune response activation, including increased CD4+ and CD8+ T cells and elevated IFN-γ levels. This study provides a promising multi-modal strategy for overcoming challenges in cancer therapy by integrating chemotherapy, immunotherapy, and targeted drug delivery, offering potential for improved treatment outcomes in ovarian cancer.
Collapse
Affiliation(s)
- Hongru Li
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, China; College of Biological and Agricultural Engineering, Jilin University, China
| | - Yuhan Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, China
| | - Shunqing Zhou
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, China
| | - Jianli Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, China
| | - Yuemei Jin
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, China.
| |
Collapse
|
4
|
Aronson SL, Thijssen B, Lopez-Yurda M, Koole SN, van der Leest P, León-Castillo A, Harkes R, Seignette IM, Sanders J, Alkemade M, Kemper I, Holtkamp MJ, Mandjes IAM, Broeks A, Lahaye MJ, Rijlaarsdam MA, van den Broek D, Wessels LFA, Horlings HM, van Driel WJ, Sonke GS. Neo-adjuvant pembrolizumab in stage IV high-grade serous ovarian cancer: the phase II Neo-Pembro trial. Nat Commun 2025; 16:3520. [PMID: 40229272 PMCID: PMC11997049 DOI: 10.1038/s41467-025-58440-y] [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: 10/22/2024] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
While immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, their efficacy in high-grade serous ovarian cancer (HGSOC) remains limited. Some patients, however, achieve lasting responses, emphasizing the need to understand how tumor microenvironment and molecular characteristics influence ICI response. The phase 2 Neo-Pembro study (NCT03126812) included 33 untreated stage IV HGSOC patients, who were scheduled for 6 cycles of carboplatin-paclitaxel and interval cytoreductive surgery. Pembrolizumab (pembro) was added from cycle two and continued for one year. The primary objective was to assess intratumoral immune activation using multiplexed immunofluorescence and immune-related gene expression. Our findings show immune activation, evidenced by an increase in CD3 + , CD8 + , CD8 + /FOXP3+ ratio, TNF-α and interferon-γ signaling. Treatment was well-tolerated. We observed major pathologic responses in 9/33 patients (27%, 95%CI 14-46), with pathologic response strongly associated with immune activation and OS. At a median follow-up of 52.8 months, 8/9 major responders were alive, with 6 patients recurrence-free. In contrast, 4/24 minor responders survived, including one recurrence-free. ctDNA clearance was observed in all major responders and was associated with prolonged PFS and OS. PD-L1 expression and homologous recombination deficiency were predictive of major response and may serve as biomarkers, warranting further exploration. These results suggest major responders may benefit from neo-adjuvant pembro.
Collapse
Affiliation(s)
- S L Aronson
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - B Thijssen
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - M Lopez-Yurda
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - S N Koole
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - P van der Leest
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A León-Castillo
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R Harkes
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - I M Seignette
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Alkemade
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - I Kemper
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M J Holtkamp
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - I A M Mandjes
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A Broeks
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M J Lahaye
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - M A Rijlaarsdam
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - D van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - L F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - H M Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - W J van Driel
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - G S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
5
|
Chalif J, Wegner L, Backes F, Chambers LM. Hyperthermic Intraperitoneal Chemotherapy in the Management of Ovarian, Fallopian Tube and Peritoneal Carcinomas. Surg Oncol Clin N Am 2025; 34:265-285. [PMID: 40015804 DOI: 10.1016/j.soc.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Ovarian cancer (OC) is a leading cause of gynecologic cancer-related deaths. Despite improvements, the 10-year survival rate has remained relatively unchanged in recent years. Disease recurrence occurs in nearly 80% of patients with advanced disease symptoms. Hyperthermic intraperitoneal chemotherapy (HIPEC) is a therapy that targets common sites of OC recurrence with heated chemotherapy during surgery, which promotes either direct cell death, heightened deoxyribonucleic acid damage, or increased tumor susceptibility to chemotherapeutics. Clinical trials utilizing HIPEC have shown promising results. This paper aims to provide an overview of HIPEC, including historical trials, mechanistic insights, recent advances, and ongoing clinical trials.
Collapse
Affiliation(s)
- Julia Chalif
- Division of Gynecologic Oncology, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Lauren Wegner
- Division of Obstetrics & Gynecology, The Ohio State University, Columbus, OH, USA
| | - Floor Backes
- Division of Gynecologic Oncology, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Laura M Chambers
- Division of Gynecologic Oncology, The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, OH, USA.
| |
Collapse
|
6
|
Wang YW, Allen I, Funingana G, Tischkowitz M, Joko-Fru YW. Predictive biomarkers for the efficacy of PARP inhibitors in ovarian cancer: an updated systematic review. BJC REPORTS 2025; 3:14. [PMID: 40069561 PMCID: PMC11897386 DOI: 10.1038/s44276-025-00122-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/06/2024] [Accepted: 01/09/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND PARP inhibitors are effective in treating ovarian cancer, especially for BRCA1/2 pathogenic variant carriers and those with HRD (homologous recombination deficiency). Concerns over toxicity and costs have led to the search for predictive biomarkers. We present an updated systematic review, expanding on a previous ESMO review on PARP inhibitor biomarkers. METHODS Following ESMO's 2020 review protocol, we extended our search to March 31, 2023, including PubMed and clinical trial data. We also reviewed the reference lists of review articles. We conducted a meta-analysis using a random-effects model to evaluate hazard ratios and assess the predictive potential of biomarkers and the effectiveness of PARP inhibitors in survival. RESULTS We found 375 articles, 103 of which were included after screening (62 primary research, 41 reviews). HRD remained the primary biomarker (95%), particularly BRCA1/2 variants (77%). In the non-HRD category, six articles (10%) introduced innovative biomarkers, including ADP-ribosylation, HOXA9 promoter methylation, patient-derived organoids, KELIM, and SLFN11. DISCUSSION Prospective assessment of real-time homologous recombination repair via nuclear RAD51 levels shows promise but needs validation. Emerging biomarkers like ADP-ribosylation, HOXA9 promoter methylation, patient-derived organoids, KELIM, and SLFN11 offer potential but require large-scale validation.
Collapse
Affiliation(s)
- Ying-Wen Wang
- Division of Gynaecologic Oncology, Department of Obstetrics and Gynaecology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| | - Isaac Allen
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Yvonne Walburga Joko-Fru
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
7
|
Melnyk M, Starczewski A, Nawrocka-Rutkowska J, Gorzko A, Melnyk B, Szydłowska I. Giant Ovarian Tumors in Young Women: Diagnostic and Treatment Challenges-A Report of Two Cases and Narrative Review of the Recent Literature. J Clin Med 2025; 14:1236. [PMID: 40004767 PMCID: PMC11856845 DOI: 10.3390/jcm14041236] [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: 12/20/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Ovarian cysts (OCs) are a common gynecological issue, with approximately 20% of women developing at least one pelvic mass during their lifetime. The incidence of large ovarian cysts has decreased substantially due to regular gynecological screenings. However, giant ovarian tumors still continue to pose significant diagnostic and therapeutic challenges. Methods: We report two cases of giant ovarian tumors (GOTs). Case 1 involves a 17-year-old woman who presented with a 2-year history of gradual abdominal enlargement, accompanied by repeated attempts at weight reduction. A computed tomography (CT) scan revealed a large tumor. It was excised by laparotomy. Histopathologic examination revealed ovarian cystadenofibroma. Case 2 presents a 25-year-old female who had a 3-month history of progressive, severe abdominal distension and weight gain, accompanied by nausea and diarrhea. CT imaging revealed a giant cystic neoplasm. The cyst was removed by laparotomy. The histopathological study revealed the intestinal-endocervical mucinous borderline tumor. In this context, we performed a narrative literature review, including cases of giant ovarian tumors in young women over the past five years. We centered on diagnoses and management in these cases. Results: The surgical management of both cases was successful, with complete tumor excision and favorable postoperative outcomes. hese cases underscore the importance of including giant ovarian tumors in the differential diagnosis of young women presenting with progressive abdominal distension. The narrative review analyzed 39 relevant publications on the management of giant ovarian tumors in young women. Conclusions: It is important to highlight a possible risk of malignancy, and risk of fatal complications during the surgical removal of giant ovarian cysts (GOCs). To ensure safer and more successful outcomes, multidisciplinary care should be provided. The early detection and diagnosis of OCs are challenging, as patients may not seek medical attention until the tumor has become large enough to cause symptoms. It is crucial to raise awareness among family doctors and other primary care providers (PCPs) regarding OCs to ensure optimal diagnostic and therapeutic management and improve the outcomes for patients with OCs.
Collapse
Affiliation(s)
- Mariia Melnyk
- Department of Gynecology, Endocrinology and Gynecologic Oncology, Pomeranian Medical University, 70-204 Szczecin, Poland
| | - Andrzej Starczewski
- Department of Gynecology, Endocrinology and Gynecologic Oncology, Pomeranian Medical University, 70-204 Szczecin, Poland
| | - Jolanta Nawrocka-Rutkowska
- Department of Gynecology, Endocrinology and Gynecologic Oncology, Pomeranian Medical University, 70-204 Szczecin, Poland
| | - Amalia Gorzko
- University Clinical Hospital No. 1, 71-252 Szczecin, Poland
| | - Bohdan Melnyk
- University Clinical Hospital No. 2, 70-111 Szczecin, Poland
| | - Iwona Szydłowska
- Department of Gynecology, Endocrinology and Gynecologic Oncology, Pomeranian Medical University, 70-204 Szczecin, Poland
| |
Collapse
|
8
|
Zelisse HS, van Gent MDJM, Mom CH, de Ridder S, Snijders MLH, Heeling M, Stoter M, Broeks A, Horlings HM, Lok CAR, Bosch SL, Piek JM, Bart J, Reyners AKL, Wisman GBA, Yigit R, Boere IA, Collée M, Groenendijk FH, Jansen MPHM, Roes EM, Hofhuis W, Hoogduin KJ, Alcalá LSM, Smedts HPM, Makkus ACF, Nieuwenhuyzen-de Boer GM, van Es N, Vencken PMLH, van Altena AM, Simons M, Hazelbag HM, Kagie MJ, Aliredjo R, Bonestroo TJJ, Bosse T, de Kroon CD, Brinkhuis M, Janssen MJ, Koster NC, Kruse AJ, Gerestein CG, Jonges TGN, Zweemer RP, Kooreman LFS, Lambrechts S, Ebisch IMW, de Kievit van der Heijden IM, Voorham QJ, van der Aa MA, Belien JAM, van de Vijver MJ, Dijk F. Evaluation of the recently established Dutch nationwide Archipelago of Ovarian Cancer Research biobank. Ann Diagn Pathol 2025; 74:152411. [PMID: 39591762 DOI: 10.1016/j.anndiagpath.2024.152411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/16/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024]
Abstract
Fundamental and translational research in ovarian cancer aims to enhance understanding of disease mechanisms and improve treatment and survival outcomes. To support this, we established the Dutch multicenter, interdisciplinary Archipelago of Ovarian Cancer Research (AOCR) infrastructure, which includes a nationwide biobank. In this study, we share our experiences in establishing the infrastructure, offer guidance for similar initiatives, and evaluate the AOCR patient cohort. Key challenges included obtaining Data Protection Impact Assessment (DPIA) clearance, drafting the consortium agreement, and securing ethical approval from all hospitals. Over three years, 1093 patients were enrolled across 17 hospitals, resulting in the collection of 1339 tissue samples and 2280 blood samples. Of the 523 patients with currently available clinical and pathological data, 74 % (n = 387) had primary ovarian cancer. Among these patients, 73.4 % was diagnosed with high-grade serous ovarian carcinoma, and 80.9 % presented with advanced-stage disease. Surgery was performed on 93 % of patients with primary ovarian cancer, and chemotherapy was administered to 90.4 % of these patients. In conclusion, the AOCR biobank has established a robust foundation for future fundamental and translational ovarian cancer research. This manuscript provides valuable insights and guidance for developing future research infrastructures and biobanks, and contains detailed information about the AOCR patient cohort to date.
Collapse
Affiliation(s)
- Hein S Zelisse
- Department of Pathology, Cancer Center Amsterdam, Amsterdam Reproduction & Development research institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Mignon D J M van Gent
- Department of Gynaecologic Oncology, Centre for Gynaecologic Oncology Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Constantijne H Mom
- Department of Gynaecologic Oncology, Centre for Gynaecologic Oncology Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Sander de Ridder
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Malou L H Snijders
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marlou Heeling
- Department of Pathology, Cancer Center Amsterdam, Amsterdam Reproduction & Development research institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Matthijs Stoter
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Annegien Broeks
- Department of CFMPB (Core Facility - Molecular Pathology and Biobanking), The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hugo M Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Christianne A R Lok
- Department of Gynaecological Oncology, Centre for Gynaecologic Oncology Amsterdam, Antoni van Leeuwenhoek - The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Steven L Bosch
- Department of Pathology, Eurofins-PAMM, Eindhoven, the Netherlands
| | - Jurgen M Piek
- Department of Obstetrics and Gynaecology, Catharina Hospital, Catharina Cancer Institute, Eindhoven, the Netherlands
| | - Joost Bart
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anna K L Reyners
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - G Bea A Wisman
- Department of Gynaecologic Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Refika Yigit
- Department of Gynaecologic Oncology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ingrid A Boere
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Margriet Collée
- Department of Clinical Genetics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Floris H Groenendijk
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Maurice P H M Jansen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Eva-Maria Roes
- Department of Gynecologic Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ward Hofhuis
- Department of Gynaecological Oncology, Franciscus Hospital, Rotterdam, the Netherlands
| | | | - Luthy S M Alcalá
- Department of Pathology, Amphia Hospital Breda, Breda, the Netherlands
| | - Huberdina P M Smedts
- Department of Obstetrics and Gynecology, Amphia Hospital, Breda, the Netherlands
| | - Alexander C F Makkus
- Department of Pathology, PAL Laboratory for Pathology Dordrecht, Dordrecht, the Netherlands
| | - Gatske M Nieuwenhuyzen-de Boer
- Department of Gynecologic Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Obstetrics and Gynaecology, Albert Schweitzer Hospital, Dordrecht, the Netherlands
| | - Nicole van Es
- Department of Pathology, Bravis Hospital, Bergen op Zoom, the Netherlands
| | - Peggy M L H Vencken
- Department of Gynecology and Obstetrics, Bravis Hospital, Bergen op Zoom, the Netherlands
| | - Anne M van Altena
- Department of Obstetrics & Gynecology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Michiel Simons
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Marjolein J Kagie
- Department of Gynaecology, Haaglanden Medical Center, The Hague, the Netherlands
| | - Riena Aliredjo
- Department of Pathology, Rijnstate Hospital, Arnhem, the Netherlands
| | - Tijmen J J Bonestroo
- Department of Gynaecology and Obstetrics, Rijnstate Hospital, Arnhem, the Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Cor D de Kroon
- Department of Obstetrics and Gynaecology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mariël Brinkhuis
- Department of Pathology, Laboratory of Pathology East Netherlands - LabPON, Hengelo, the Netherlands
| | - Marc-Jan Janssen
- Department of Gynecological Oncology, Medical Spectrum Twente, Enschede, the Netherlands
| | - Nils C Koster
- Department of Pathology, Isala Clinics, Zwolle, the Netherlands
| | - Arnold-Jan Kruse
- Department of Obstetrics and Gynaecology, Isala Clinics, Zwolle, the Netherlands
| | - Cornelis G Gerestein
- Department of Gynaecological Oncology, Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Trudy G N Jonges
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ronald P Zweemer
- Department of Gynaecological Oncology, Division of Imaging and Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Loes F S Kooreman
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Sandrina Lambrechts
- Department of Obstetrics and Gynaecology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Inge M W Ebisch
- Department of Obstetrics and Gynaecology, Canisius Wilhelmina Hospital, Nijmegen, the Netherlands
| | | | | | - Maaike A van der Aa
- Department of Research and Development, IKNL - Netherlands Comprehensive Cancer Organisation, Utrecht, the Netherlands
| | - Jeroen A M Belien
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Marc J van de Vijver
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Frederike Dijk
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Vledder A, Paijens ST, Loiero D, Maagdenberg A, Duiker EW, Bart J, Hendriks AM, Jalving M, Werner N, van Rooij N, Plat A, Wisman GBA, Yigit R, Roelofsen T, Kruse AJ, de Lange NM, Koelzer VH, de Bruyn M, Nijman HW. B cells critical for outcome in high grade serous ovarian carcinoma. Int J Cancer 2024; 155:2265-2276. [PMID: 39175107 DOI: 10.1002/ijc.35149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 08/24/2024]
Abstract
Recent work has shown evidence for the prognostic significance of tumor infiltrating B cells (B-TIL) in high grade serous ovarian carcinoma (HGSOC), the predominant histological subtype of ovarian cancer. However, it remains unknown how the favorable prognosis associated with B-TIL relates to the current standard treatments of primary debulking surgery (PDS) followed by chemotherapy or (neo-)adjuvant chemotherapy (NACT) combined with interval debulking surgery. To address this, we analyzed the prognostic impact of B-TIL in relationship to primary treatment and tumor infiltrating T cell status in a highly homogenous cohort of HGSOC patients. This analysis involved a combined approach utilizing histological data and high-dimensional flow cytometry analysis. Our findings indicate that while HGSOC tumors pre-treated with NACT are infiltrated with tumor-reactive CD8+ and CD4+ TIL subsets, only B-TIL and IgA plasma blasts confer prognostic benefit in terms of overall survival. Importantly, the prognostic value of B-TIL and IgA plasma blasts was not restricted to patients treated with NACT, but was also evident in patients treated with PDS. Together, our data point to a critical prognostic role for B-TIL in HGSOC patients independent of T cell status, suggesting that alternative treatment approaches focused on the activation of B cells should be explored for HGSOC.
Collapse
Affiliation(s)
- Annegé Vledder
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sterre T Paijens
- Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dominik Loiero
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zürich, Switzerland
| | - Alexis Maagdenberg
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Evelien W Duiker
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Joost Bart
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anne M Hendriks
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Naomi Werner
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nienke van Rooij
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annechien Plat
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G Bea A Wisman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Refika Yigit
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Thijs Roelofsen
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Arnold J Kruse
- Department of Obstetrics and Gynecology, Isala Hospital Zwolle, Zwolle, The Netherlands
| | - Nastascha M de Lange
- Department of Obstetrics and Gynecology, Isala Hospital Zwolle, Zwolle, The Netherlands
| | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zürich, Switzerland
| | - Marco de Bruyn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hans W Nijman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
10
|
Kozłowska E, Haltia UM, Puszynski K, Färkkilä A. Mathematical modeling framework enhances clinical trial design for maintenance treatment in oncology. Sci Rep 2024; 14:29721. [PMID: 39613825 DOI: 10.1038/s41598-024-80768-6] [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: 06/21/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024] Open
Abstract
Clinical trials are costly and time-intensive endeavors, with a high rate of drug candidate failures. Moreover, the standard approaches often evaluate drugs under a limited number of protocols. In oncology, where multiple treatment protocols can yield divergent outcomes, addressing this issue is crucial. Here, we present a computational framework that simulates clinical trials through a combination of mathematical and statistical models. This approach offers a means to explore diverse treatment protocols efficiently and identify optimal strategies for oncological drug administration. We developed a computational framework with a stochastic mathematical model as its core, capable of simulating virtual clinical trials closely recapitulating the clinical scenarios. Testing our framework on the landmark SOLO-1 clinical trial investigating Poly-ADP-Ribose Polymerase maintenance treatment in high-grade serous ovarian cancer, we demonstrate that managing toxicity through treatment interruptions or dose reductions does not compromise treatment's clinical benefits. Additionally, we provide evidence suggesting that further reduction of hematological toxicity could significantly improve the clinical outcomes. The value of this computational framework lies in its ability to expedite the exploration of new treatment protocols, delivering critical insights pivotal to shaping the landscape of upcoming clinical trials.
Collapse
Affiliation(s)
- Emilia Kozłowska
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, 44-100, Gliwice, Poland
| | - Ulla-Maija Haltia
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Krzysztof Puszynski
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, 44-100, Gliwice, Poland.
| | - Anniina Färkkilä
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland.
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Sciences, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine, Helsinki, Finland.
| |
Collapse
|
11
|
Wang D, Zhang S, Wang Q, Li P, Liu Y. Circ_0001741 exerts as a tumor promoter in ovarian cancer through the regulation of miR-491-5p/PRSS8 axis. Discov Oncol 2024; 15:643. [PMID: 39527152 PMCID: PMC11554978 DOI: 10.1007/s12672-024-01474-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) are important regulators for ovarian cancer (OC). Circ_0001741 has been found to be highly expressed in OC samples and is involved in regulating paclitaxel resistance in OC cells. Therefore, circ_0001741 may play a vital role in OC process, and its potential molecular mechanism is worth further revealing. METHODS Circ_0001741, miR-491-5p, and PRSS8 levels in OC tumor tissues and cells were quantified by quantitative real-time PCR or western blot. The proliferation, apoptosis and metastasis of OC cells were detected by cell counting kit 8 assay, Edu assay, flow cytometry, and transwell assay. RNA interaction was verified by dual-luciferase reporter assay and RIP assay. Xenograft assay was used to detect the effect of circ_0001741 knockdown on OC tumor growth in vivo. RESULTS Circ_0001741 was upregulated in OC tissues and cell lines. Knockdown of circ_0001741 repressed OC cell proliferation, metastasis, and enhanced apoptosis. Mechanistically, miR-491-5p was targeted by circ_0001741, and miR-491-5p inhibitor could attenuate the effect of circ_0001741 silencing on OC cell progression. Meanwhile, PRSS8 was a target of miR-491-5p, and miR-491-5p overexpression inhibited OC cell progression by targeting PRSS8. Circ_0001741 regulated PRSS8 expression by sponging miR-491-5p. Besides, circ_0001741 knockdown also inhibited OC tumor growth in vivo. CONCLUSION Our data showed that circ_0001741 could promote the growth and metastasis of OC cells through the miR-491-5p/PRSS8 axis, which provided a potential molecular target for the treatment of OC.
Collapse
Affiliation(s)
- Ding Wang
- Department of Gynaecology, Yi Chang Maternal and Child Health Hospital, 99 Chengdong Avenue, Wujiagang District, Yichang City, 443001, Hubei, China.
| | - Sumin Zhang
- Department of Gynaecology, Yi Chang Maternal and Child Health Hospital, 99 Chengdong Avenue, Wujiagang District, Yichang City, 443001, Hubei, China
| | - Qiaoling Wang
- Department of Gynaecology, Yi Chang Maternal and Child Health Hospital, 99 Chengdong Avenue, Wujiagang District, Yichang City, 443001, Hubei, China
| | - Pengrong Li
- Department of Gynaecology, Yi Chang Maternal and Child Health Hospital, 99 Chengdong Avenue, Wujiagang District, Yichang City, 443001, Hubei, China
| | - Yunxia Liu
- Department of Gynaecology, Yi Chang Maternal and Child Health Hospital, 99 Chengdong Avenue, Wujiagang District, Yichang City, 443001, Hubei, China
| |
Collapse
|
12
|
Tao T, Zhang Y, Guan C, Wang S, Liu X, Wang M. Ubiquitin Ligase TRIM22 Inhibits Ovarian Cancer Malignancy via TCF4 Degradation. Mol Cancer Res 2024; 22:943-956. [PMID: 38842601 DOI: 10.1158/1541-7786.mcr-23-0962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/27/2023] [Accepted: 06/04/2024] [Indexed: 06/07/2024]
Abstract
Ovarian cancer is one of the most common malignancies in women. Tripartite motif-containing protein 22 (TRIM22) plays an important role in the initiation and progression of malignant tumors. Similarly, the transcription factor 4 (TCF4) is an essential factor involved in the initiation and progression of many tumors. However, it is still unclear whether TRIM22 can affect TCF4 in ovarian cancer. Therefore, this study aims to investigate the mechanism related to TRIM22 and TCF4 in ovarian cancer. TRIM22 protein and mRNA levels were analyzed in samples from clinical and cell lines. The effects of TRIM22 knockdown and overexpression on cell proliferation, colony formation, migration, invasion, and related biomarkers were evaluated. In addition, the role of ubiquitination-mediated degradation of TCF4 was investigated by qRT-PCR and Western blotting. The association between TRIM22 and TCF4 was evaluated by Western blotting, coimmunoprecipitation, proliferation, colony formation, invasion, migration, and related biomarkers. The results showed that the expression of TRIM22 was minimal in ovarian cancer tissues. Furthermore, upregulation of TRIM22 significantly inhibited ovarian cancer cell proliferation, colony formation, migration, and invasion. In addition, TRIM22 was observed to regulate the degradation of TCF4 through the ubiquitination pathway. TCF4 can reverse the effects of TRIM22 on proliferation, colony formation, migration, and invasion in ovarian cancer cells. TRIM22-mediated ubiquitination of TCF4 at K48 is facilitated by the RING domain. Implications: In conclusion, ubiquitination of TCF4 protein in ovarian cancer is regulated by TRIM22, which has the potential to limit the proliferation, migration, and invasion of ovarian cancer.
Collapse
Affiliation(s)
- Tao Tao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yongqi Zhang
- Department of Gynecology, The Red Cross Center Hospital of Harbin, Harbin, China
| | | | - Shuxiang Wang
- Department of Gynecology, The Red Cross Center Hospital of Harbin, Harbin, China
| | - Xiaoli Liu
- Department of Gynecology, The Red Cross Center Hospital of Harbin, Harbin, China
| | - Min Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Yeh TH, Wu CH, Ou YC, Fu HC, Lin H. A nomogram to predict platinum-sensitivity and survival outcome in women with advanced epithelial ovarian cancer. Taiwan J Obstet Gynecol 2024; 63:709-716. [PMID: 39266152 DOI: 10.1016/j.tjog.2024.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2024] [Indexed: 09/14/2024] Open
Abstract
OBJECTIVE This study presents the development and validation of a nomogram aimed at predicting platinum-sensitivity and survival outcomes in women with advanced epithelial ovarian cancer (EOC). MATERIALS AND METHODS Data from a retrospective cohort of women diagnosed with stage III/IV EOC between Jan 2011 and Dec 2021 treated at our institute were collected. Clinical and pathological characteristics were analyzed using logistic regression analysis to identify independent predictors of platinum-sensitivity. Impact on progression-free (PFS) and overall survival (OS) was determined by Kaplan-Meier and Cox regression analysis. A nomogram was constructed based on the significant predictors, and its performance was evaluated using calibration, discrimination, and validation analyses. RESULTS Of the 210 patients, 139 (66.19%) had platinum-sensitive and 71 (33.81%) were platinum-resistant disease. On multivariate analysis, platinum-resistance correlated with neoadjuvant chemotherapy (OR 2.15; 95% CI 1.10-4.21), clear cell/mucinous histology (OR 5.04; 95% CI 2.20-11.54), and sub-optimal debulking status (OR 3.37; 95% CI 1.44-7.91). Median PFS and OS were also significantly shorter for patients with neoadjuvant chemotherapy (23 vs. 10 months and 69 vs. 29 months, respectively), clear cell/mucinous histology (15 vs. 3 months and 63 vs. 11 months, respectively), and suboptimal debulking (26 vs. 5 months and 78 vs. 24 months, respectively). The nomogram demonstrated good predictive accuracy for platinum-sensitivity in the cohort as indicated by high concordance index of 0.745. Calibration plots showed excellent agreement and internal validation further confirmed the reliability of the nomogram's performance. CONCLUSION A novel predictive nomogram based on type of initial treatment, histology, and debulking status was developed, which provides a friendly and reliable tool for predicting platinum-sensitivity and survival outcomes in women with advanced EOC. Its application may assist clinicians in individualizing treatment decisions.
Collapse
Affiliation(s)
- Tsung-Hsin Yeh
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chen-Hsuan Wu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Che Ou
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Obstetrics and Gynecology, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
| | - Hung-Chun Fu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hao Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
14
|
Hemminki K, Zitricky F, Försti A, Hemminki A. Survival in Elderly Ovarian Cancer Remains Challenging in the Nordic Countries. Cancers (Basel) 2024; 16:2198. [PMID: 38927904 PMCID: PMC11201377 DOI: 10.3390/cancers16122198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Despite treatment having improved through intensive surgical procedures and chemotherapy-and more recently, targeted therapies-ovarian cancer is the most fatal female cancer. As such, we wanted to analyze age-specific survival trends for ovarian cancer in Denmark, Finland, Norway and Sweden over the past 50 years, with a special aim of comparing survival development between the age groups. METHODS We modelled survival data from the NORDCAN database for 1-, 5- and conditional 5/1-year relative (between years 1 and 5) survival for ovarian cancer from 1972 to 2021. RESULTS Young patients had a 70% 5-year survival while the survival was only 30% for the oldest patients. Conditional survival showed that survival between years 1 and 5 did not improve for patients older than 60 years throughout the 50-year period, during which time the gaps between the youngest and the oldest patients widened. CONCLUSIONS Improvement in 1-year survival was so large that it masked the modest development between years 1 and 5, resulting in a widening age disparity in 5-year survival. The current treatment practices, which appear increasingly effective for younger patients, have not helped remedy the large age differences in ovarian cancer survival. Early detection methods and therapeutic innovations are urgently needed, and aged patients need a special focus.
Collapse
Affiliation(s)
- Kari Hemminki
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, 30605 Pilsen, Czech Republic;
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany
| | - Frantisek Zitricky
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, 30605 Pilsen, Czech Republic;
| | - Asta Försti
- Hopp Children’s Cancer Center (KiTZ), 69120 Heidelberg, Germany;
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland;
- Comprehensive Cancer Center, Helsinki University Hospital, 00290 Helsinki, Finland
| |
Collapse
|
15
|
Duan C, Yan Z, Wu C, Zhou X, Bao W. DNA methylation characteristics associated with chemotherapy resistance in epithelial ovarian cancer. Heliyon 2024; 10:e27212. [PMID: 38468944 PMCID: PMC10926131 DOI: 10.1016/j.heliyon.2024.e27212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
Objective The high mortality rate of epithelial ovarian cancer (EOC) is often attributed to the frequent development of chemoresistance. DNA methylation is a predictive biomarker for chemoresistance. Methods This study utilized DNA methylation profiles and relevant information from GEO and TCGA to identify different methylated CpG sites (DMCs) between chemoresistant and chemosensitive patients. Subsequently, we constructed chemoresistance risk models with DMCs. The genes corresponding to candidate DMCs in chemoresistance risk models were further analyzed to identify different methylated gene symbols (DMGs) associated with chemoresistance. The DMGs that showed a strong correlation with the corresponding DMCs were analyzed through immunohistochemistry. Results Compared to chemosensitive EOC patients, chemoresistant patients showed 423 hypermethylated CpGs and 1445 hypomethylated CpGs. The chemoresistance risk models based on DMCs have shown the improved predictive ability for chemoresistance in EOC (AUC = 65.0-76.2%). The methylations of cg25510164, cg13154880, cg15362155 and cg08665359 were strongly associated with decreased risk of chemoresistance. Conversely, the methylation of cg08872590 and cg14739437 significantly increased the risk. We identified 13 DMGs, from 47 DMCs corresponding genes, between chemosensitive and chemoresistant samples. Among the DMGs, the expression levels of DDR2 and OPCML exhibited strong correlations with the corresponding DMCs. DDR2 and OPCML both showed enhanced expression in chemoresistant ovarian microarray tissue. Conclusions Hypomethylated CpGs may play a significant role in DNA methylation associated with chemoresistance in EOC. The epigenetic modification of DDR2 could have important implications for the development of chemoresistance. Our study provides valuable insights for future research on DNA methylation in the chemoresistance of EOC.
Collapse
Affiliation(s)
| | | | - Cailiang Wu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080, Shanghai, China
| | - Xuexin Zhou
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080, Shanghai, China
| | - Wei Bao
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 200080, Shanghai, China
| |
Collapse
|
16
|
van der Ploeg P, Hendrikse CSE, Thijs AMJ, Westgeest HM, Smedts HPM, Vos MC, Jalving M, Lok CAR, Boere IA, van Ham MAPC, Ottevanger PB, Westermann AM, Mom CH, Lalisang RI, Lambrechts S, Bekkers RLM, Piek JMJ. Phenotype-guided targeted therapy based on functional signal transduction pathway activity in recurrent ovarian cancer patients: The STAPOVER study protocol. Heliyon 2024; 10:e23170. [PMID: 38187310 PMCID: PMC10770441 DOI: 10.1016/j.heliyon.2023.e23170] [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] [Received: 04/19/2023] [Revised: 11/15/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024] Open
Abstract
Objective Ovarian cancer is the fifth cause of cancer-related death among women. The benefit of targeted therapy for ovarian cancer patients is limited even if treatment is stratified by molecular signature. There remains a high unmet need for alternative diagnostics that better predict targeted therapy, as current diagnostics are generally inaccurate predictors. Quantitative assessment of functional signal transduction pathway (STP) activity from mRNA measurements of target genes is an alternative approach. Therefore, we aim to identify aberrantly activated STPs in tumour tissue of patients with recurrent ovarian cancer and start phenotype-guided targeted therapy to improve survival without compromising quality of life. Study design Patients with recurrent ovarian cancer and either 1) have platinum-resistant disease, 2) refrain from standard therapy or 3) are asymptomatic and not yet eligible for standard therapy will be included in this multi-centre prospective cohort study with multiple stepwise executed treatment arms. Targeted therapy will be available for patients with aberrantly high functional activity of the oestrogen receptor, androgen receptor, phosphoinositide 3-kinase or Hedgehog STP. The primary endpoint of this study is the progression-free survival (PFS) ratio (PFS2/PFS1 ratio) according to RECIST 1.1 determined by the PFS on matched targeted therapy (PFS2) compared to PFS on prior therapy (PFS1). Secondary endpoints include among others best overall response, overall survival, side effects, health-related quality of life and cost-effectiveness. Conclusion The results of this study will show the clinical applicability of STP activity in selecting recurrent ovarian cancer patients for effective therapies.
Collapse
Affiliation(s)
- Phyllis van der Ploeg
- Department of Obstetrics and Gynaecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Cynthia SE. Hendrikse
- Department of Obstetrics and Gynaecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Anna MJ. Thijs
- Department of Internal Medicine and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
| | - Hans M. Westgeest
- Department of Internal Medicine, Amphia Hospital, Breda, the Netherlands
| | - Huberdina PM. Smedts
- Department of Obstetrics and Gynaecology, Amphia Hospital, Breda, the Netherlands
| | - M Caroline Vos
- Department of Obstetrics and Gynaecology, Elisabeth-Tweesteden Hospital, Tilburg, the Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Christianne AR. Lok
- Department of Gynaecologic Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Ingrid A. Boere
- Department of Medical Oncology, Erasmus Medical Centre Cancer Institute, Rotterdam, the Netherlands
| | - Maaike APC. van Ham
- Department of Obstetrics and Gynaecology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | | | - Anneke M. Westermann
- Department of Oncology, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Constantijne H. Mom
- Department of Obstetrics and Gynaecology, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Roy I. Lalisang
- Department of Medical Oncology, Maastricht University Medical Centre +, Maastricht, the Netherlands
| | - Sandrina Lambrechts
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre +, Maastricht, the Netherlands
| | - Ruud LM. Bekkers
- Department of Obstetrics and Gynaecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Jurgen MJ. Piek
- Department of Obstetrics and Gynaecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
| |
Collapse
|
17
|
Chang YH, Wu KC, Wang KH, Ding DC. Ovarian Cancer Patient-Derived Organoids Used as a Model for Replicating Genetic Characteristics and Testing Drug Responsiveness: A Preliminary Study. Cell Transplant 2024; 33:9636897241281869. [PMID: 39323050 PMCID: PMC11425734 DOI: 10.1177/09636897241281869] [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] [Revised: 12/29/2023] [Accepted: 08/21/2024] [Indexed: 09/27/2024] Open
Abstract
This study aimed to explore the role of ovarian cancer patient-derived organoids (PDOs) in their replicating genetic characteristics and testing drug responsiveness. Ovarian cancer PDOs were cultured in Matrigel with a specialized medium. The successful rate and proliferation rate were calculated. Morphology, histology, and immunohistochemistry (IHC) (PAX8, P53, and WT1) were used to identify the tumor characteristics. Gene sequencing, variant allele frequency (VAF), and copy number variation were used to explore the mutation profile. The sensitivity to chemodrugs (carboplatin, paclitaxel, gemcitabine, doxorubicin, and olaparib) was conducted. Successful generation of organoids occurred in 54% (7/13) of attempts, encompassing 4 high-grade serous carcinomas (HGSC), 1 mucinous carcinoma (MC), 1 clear cell carcinoma (CCC), and 1 carcinosarcoma. The experiments used six organoids (3 HGSC, 1 CCC, 1 MC, and 1 carcinosarcoma). The derived organoids exhibited spherical-like morphology, and the diameter ranged from 100 to 500 μm. The histology and IHC exhibited the same between organoids and primary tumors. After cryopreservation, the organoid's growth rate was slower than the primary culture (14 days vs 10 days, P < 0.01). Targeted sequencing revealed shared DNA variants, including mutations in key genes, such as BRCA1, PIK3CA, ARID1A, and TP53. VAF was similar between primary tumors and organoids. The organoids maintained inherited most copy number alterations. Drug sensitivity testing revealed varying responses, with carcinosarcoma organoids showing higher sensitivity to paclitaxel and gemcitabine than HGSC organoids. Our preliminary results showed that ovarian cancer PDOs could be successfully derived and histology, mutations, and diverse copy numbers of genotypes could be faithfully captured. Drug testing could reveal the individual PDO's responsiveness to drugs. PDOs might be as valuable resources for investigating genomic biomarkers for personalized treatment.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien
| | - Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien
| | - Kai-Hung Wang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien
- Institute of Medical Sciences, College of Medicine, Tzu Chi University, Hualien
| |
Collapse
|
18
|
Fetsych M, Igumentseva N, Ferneza S, Yarema R, Volodko N. CYTOKINE-MODULATING INFLUENCE OF HIPEC ON THE INTRAPERITONEAL HOMEOSTASIS FORMATION IN PATIENTS WITH OVARIAN CANCER. PROCEEDING OF THE SHEVCHENKO SCIENTIFIC SOCIETY. MEDICAL SCIENCES 2023; 72. [DOI: 10.25040/ntsh2023.02.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/14/2024]
Abstract
Background: Ovarian cancer (OC) has the greatest mortality rate among oncogynecological diseases. Most cases are diagnosed at the peritoneal dissemination stage, resulting in radical treatment. Most cases (75%) are diagnosed in III-IV stages at the stage of peritoneal dissemination, making the radical treatment impossible. Intraoperative hyperthermic intraperitoneal chemoperfusion (HIPEC), which has ceased to be an experimental technique over the past decade, is increasingly used among the methods of combating peritoneal carcinomatosis and demonstrates an increase in recurrence-free and overall survival in advanced stages of OC. Nevertheless, certain pathophysiological aspects of the effect of HIPEC on intra-abdominal homeostasis, and therefore on the further course of the disease, have not been elucidated. However, understanding this effect may be the key to the successful application of HIPEC and predicting its efficacy in each case of OC.
Objectives: To access changes in intraperitoneal homeostasis in patients with advanced OC after HIPEC procedure (as a stage of cytoreductive surgery); in particular, to evaluate changes of TNF and TGF-β expression under the influence of HIPEC and systemic chemotherapy in the OC microenvironment.
Materials and methods: The study included 33 OC patients treated at Lviv Regional Cancer Centre in 2016–2020. Twelve of them received HIPEC. The primary tumor cultures and primary cultures of peritoneal mesothelium and macrophages were obtained for all patients. The cytokines TNF and TGF-β activity were determined in their conditional media.
Results: The decrease of TNF and TGF-β concentrations in the drainage exudate after HIPEC was revealed compared with the same parameters in the ascitic fluid before the operation. One-hour hyperthermia of the primary culture of peritoneal ascites-associated macrophages of recurrent OC patients decreased the TNF level in conditional media. A decline was found in 10 out of 12 cases.
Conclusions: HIPEC has a cytokine-modulating effect on the intra-abdominal homeostasis of patients with OC, suppressing TNF expression by peritoneal macrophages. Reduced activity of TNF in supernatants of OC primary tumor cultures was associated with higher sensitivity to chemotherapy.
Collapse
|
19
|
Farokhi Boroujeni S, Rodriguez G, Galpin K, Yakubovich E, Murshed H, Ibrahim D, Asif S, Vanderhyden BC. BRCA1 and BRCA2 deficient tumour models generate distinct ovarian tumour microenvironments and differential responses to therapy. J Ovarian Res 2023; 16:231. [PMID: 38017453 PMCID: PMC10683289 DOI: 10.1186/s13048-023-01313-z] [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: 08/18/2023] [Accepted: 11/09/2023] [Indexed: 11/30/2023] Open
Abstract
Clinical trials are currently exploring combinations of PARP inhibitors and immunotherapies for the treatment of ovarian cancer, but their effects on the ovarian tumour microenvironment (TME) remain unclear. Here, we investigate how olaparib, PD-L1 monoclonal antibodies, and their combination can influence TME composition and survival of tumour-bearing mice. We further explored how BRCA deficiencies can influence the response to therapy. Olaparib and combination therapies similarly improved the median survival of Brca1- and Brca2-deficient tumour-bearing mice. Anti-PD-L1 monotherapy improved the survival of mice with Brca1-null tumours, but not Brca2-null tumours. A detailed analysis of the TME revealed that olaparib monotherapy resulted in a large number of immunosuppressive and immunomodulatory effects in the more inflamed Brca1-deficient TME but not Brca2-deficient tumours. Anti-PD-L1 treatment was mostly immunosuppressive, resulting in a systemic reduction of cytokines and a compensatory increase in PD-L1 expression. The results of the combination therapy generally resembled the effects of one or both of the monotherapies, along with unique changes observed in certain immune populations. In-silico analysis of RNA-seq data also revealed numerous differences between Brca-deficient tumour models, such as the expression of genes involved in inflammation, angiogenesis and PD-L1 expression. In summary, these findings shed light on the influence of novel therapeutics and BRCA mutations on the ovarian TME.
Collapse
Affiliation(s)
- Salar Farokhi Boroujeni
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Galaxia Rodriguez
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Kristianne Galpin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Edward Yakubovich
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Humaira Murshed
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Dalia Ibrahim
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Sara Asif
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Barbara C Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
20
|
Mazidimoradi A, Momenimovahed Z, Khani Y, Rezaei Shahrabi A, Allahqoli L, Salehiniya H. Global patterns and temporal trends in ovarian cancer morbidity, mortality, and burden from 1990 to 2019. ONCOLOGIE 2023; 25:641-659. [DOI: 10.1515/oncologie-2023-0172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Objectives
Ovarian cancer (OC) is the deadliest gynecological cancer in the world. Deeper knowledge over time is the basis for global studies to design and implement effective measures to reduce inequalities; this study was conducted to investigate the trend of OC incidence and management in the world from 1990 to 2019.
Methods
We obtained crude numbers and age-standardized rate (ASRs) of OC annually from the 2019 Global Burden of Disease (GBD) study to examine OC’s morbidity, mortality rates, and disability-adjusted life years (DALYs) based on age group, sociodemographic index (SDI), WHO regions, continents, World Bank regions, and GBD regions from 1990 to 2019 in 204 countries and territories. The relative change (%) and average Annual Percent Change (AAPC) were used to display the epidemiological trend.
Results
Globally, the number of OC incidents increased from 141,706 in 1990 to 294,422 in 2019. Despite the age-standardized incidence rate (ASIR) in regions with high SDI having a downward trend, these areas recorded the highest incidence cases and ASIR in 2019. Although the World Bank high-income level had the most frequent incidence cases and ASIR, the ASIR in these regions decreased from 1990 to 2019. Among the continents, Europe and America have the highest ASIR but experienced a decreasing trend from 1990 until 2019 in ASIR. The age-standardized mortality rate (ASMR) in the World Bank high-income level experienced a decreasing trend in 1990–2019. In contrast, in the middle, low-middle, and low SDI regions, the death number increased approximately 3.5–4.1 times, and the ASMR had a significant increase from 0.5 in the middle to 0.75 in the low-middle SDI. Globally, the DALY cases of OC rose from 2,732,666 in 1990 to 5,359,737 in 2019; almost doubling. A significant decrease in the DALYs ASR was observed in seven GBD regions. The most pronounced decrease was found in Australia.
Conclusions
The trend of OC incidence and burden and approximate mortality were stable from 1990 to 2019; especially in lower socioeconomic areas and low-income countries; while the incidence ASR of this cancer in the high SDI regions decreased from 1990 to 2019. The key to reducing OC remains in primary prevention. Approaches such as weight loss, a healthy lifestyle and diet, promoting childbearing and breastfeeding, and recommending the use of oral contraceptives in eligible individuals can have a protective effect against this silent killer.
Collapse
Affiliation(s)
- Afrooz Mazidimoradi
- Health Assistant Department , Shiraz University of Medical Sciences , Shiraz , Iran
| | | | - Yousef Khani
- Clinical Research Development Unit, Shahid Madani Hospital , Alborz University of Medical Sciences , Karaj , Iran
- Department of Epidemiology, School of Public Health and Safety , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | | | - Leila Allahqoli
- Midwifery Department , Ministry of Health and Medical Education , Tehran , Iran
| | - Hamid Salehiniya
- Department of Epidemiology and Biostatistics, School of Health, Social Determinants of Health Research Center , Birjand University of Medical Sciences , Birjand , Iran
| |
Collapse
|
21
|
Climent MT, Serra A, Balaguer C, Llueca A. Should We Abandon Intraperitoneal Chemotherapy in the Treatment of Advanced Ovarian Cancer? A Meta-Analysis. J Pers Med 2023; 13:1636. [PMID: 38138863 PMCID: PMC10745120 DOI: 10.3390/jpm13121636] [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: 10/17/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Ovarian cancer is the gynaecological malignancy with the highest mortality and diagnosis often occurs in its advanced stages. Standard treatment in these cases is based on complete cytoreductive surgery with adjuvant intravenous chemotherapy. Other types of treatment are being evaluated to improve the prognosis of these patients, including intraperitoneal chemotherapy and antiangiogenic therapy. These may improve survival or time to relapse in addition to intravenous chemotherapy. OBJECTIVE The aim of this meta-analysis is to determine whether treatment with intravenous chemotherapy remains the gold standard, or whether the addition of intraperitoneal chemotherapy has a benefit in overall survival (OS) and disease-free interval (DFS). MATERIALS AND METHODS A literature search was carried out in Pubmed and Cochrane, selecting clinical studies and systematic reviews published in the last 10 years. Statistical analysis was performed using the hazard ratio measure in the RevMan tool. RESULTS Intraperitoneal chemotherapy shows a benefit in OS and DFS compared with standard intravenous chemotherapy. The significant differences in OS (HR: 0.81 CI 95% 0.74-0.88) and in DFS (HR: 0.81 CI 95% 0.75-0.87) are statistically significant (p < 0.00001). There were no clinical differences in toxicity and side-effects. CONCLUSION Intraperitoneal chemotherapy is an option that improves OS and DFS without significant toxicity regarding the use of intravenous chemotherapy alone. However, prospective studies are needed to determine the optimal dose and treatment regimen that will maintain the benefits while minimising side effects and toxicity and the profile of patients who will benefit most from this treatment.
Collapse
Affiliation(s)
- Maria Teresa Climent
- Multidisciplinary Unit of Abdominopelvic Oncological Surgery (MUAPOS), Department of Obstetrics and Gynaecology, Hospital General Universitario de Castellón, 12004 Castellón, Spain; (A.S.); (A.L.)
- Oncological Surgery Research Group (OSRG), Department of Medicine, University Jaume I (UJI), 12004 Castellón, Spain
| | - Anna Serra
- Multidisciplinary Unit of Abdominopelvic Oncological Surgery (MUAPOS), Department of Obstetrics and Gynaecology, Hospital General Universitario de Castellón, 12004 Castellón, Spain; (A.S.); (A.L.)
- Oncological Surgery Research Group (OSRG), Department of Medicine, University Jaume I (UJI), 12004 Castellón, Spain
| | - Carolina Balaguer
- Department of Medicine, University Jaume I (UJI), 12004 Castellón, Spain;
| | - Antoni Llueca
- Multidisciplinary Unit of Abdominopelvic Oncological Surgery (MUAPOS), Department of Obstetrics and Gynaecology, Hospital General Universitario de Castellón, 12004 Castellón, Spain; (A.S.); (A.L.)
- Oncological Surgery Research Group (OSRG), Department of Medicine, University Jaume I (UJI), 12004 Castellón, Spain
| |
Collapse
|
22
|
Åkerlund E, Gudoityte G, Moussaud-Lamodière E, Lind O, Bwanika HC, Lehti K, Salehi S, Carlson J, Wallin E, Fernebro J, Östling P, Kallioniemi O, Joneborg U, Seashore-Ludlow B. The drug efficacy testing in 3D cultures platform identifies effective drugs for ovarian cancer patients. NPJ Precis Oncol 2023; 7:111. [PMID: 37907613 PMCID: PMC10618545 DOI: 10.1038/s41698-023-00463-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 10/06/2023] [Indexed: 11/02/2023] Open
Abstract
Most patients with advanced ovarian cancer (OC) relapse and progress despite systemic therapy, pointing to the need for improved and tailored therapy options. Functional precision medicine can help to identify effective therapies for individual patients in a clinically relevant timeframe. Here, we present a scalable functional precision medicine platform: DET3Ct (Drug Efficacy Testing in 3D Cultures), where the response of patient cells to drugs and drug combinations are quantified with live-cell imaging. We demonstrate the delivery of individual drug sensitivity profiles in 20 samples from 16 patients with ovarian cancer in both 2D and 3D culture formats, achieving over 90% success rate in providing results six days after operation. In this cohort all patients received carboplatin. The carboplatin sensitivity scores were significantly different for patients with a progression free interval (PFI) less than or equal to 12 months and those with more than 12 months (p < 0.05). We find that the 3D culture format better retains proliferation and characteristics of the in vivo setting. Using the DET3Ct platform we evaluate 27 tailored combinations with results available 10 days after operation. Notably, carboplatin and A-1331852 (Bcl-xL inhibitor) showed an additive effect in four of eight OC samples tested, while afatinib and A-1331852 led to synergy in five of seven OC models. In conclusion, our 3D DET3Ct platform can rapidly define potential, clinically relevant data on efficacy of existing drugs in OC for precision medicine purposes, as well as provide insights on emerging drugs and drug combinations that warrant testing in clinical trials.
Collapse
Affiliation(s)
- Emma Åkerlund
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Greta Gudoityte
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | | | - Olina Lind
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | | | - Kaisa Lehti
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology NTNU, Trondheim, Norway
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sahar Salehi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden
| | - Joseph Carlson
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Emelie Wallin
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Josefin Fernebro
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden
- Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Päivi Östling
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Olli Kallioniemi
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Ulrika Joneborg
- Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden
| | - Brinton Seashore-Ludlow
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
23
|
Aronson SL, Lopez-Yurda M, Koole SN, Schagen van Leeuwen JH, Schreuder HWR, Hermans RHM, de Hingh IHJT, van Gent MDJM, Arts HJG, van Ham MAPC, van Dam PA, Vuylsteke P, Aalbers AGJ, Verwaal VJ, Van de Vijver KK, Aaronson NK, Sonke GS, van Driel WJ. Cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy in patients with advanced ovarian cancer (OVHIPEC-1): final survival analysis of a randomised, controlled, phase 3 trial. Lancet Oncol 2023; 24:1109-1118. [PMID: 37708912 DOI: 10.1016/s1470-2045(23)00396-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/25/2023] [Accepted: 08/04/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND The OVHIPEC-1 trial previously showed that the addition of hyperthermic intraperitoneal chemotherapy (HIPEC) to interval cytoreductive surgery resulted in improved progression-free and overall survival compared with cytoreductive surgery alone at 4·7 years of follow-up in patients with stage III epithelial ovarian cancer who were ineligible for primary cytoreduction. We report the final survival outcomes after 10 years of follow-up. METHODS In this open-label, randomised, controlled, phase 3 trial, patients with primary epithelial stage III ovarian cancer were recruited at eight HIPEC centres in the Netherlands and Belgium. Patients were eligible if they were aged 18-76 years, had not progressed during at least three cycles of neoadjuvant carboplatin plus paclitaxel, had a WHO performance status score of 0-2, normal blood counts, and adequate renal function. Patients were randomly assigned (1:1) to undergo interval cytoreductive surgery without HIPEC (surgery group) or with HIPEC (100 mg/m2 cisplatin; surgery-plus-HIPEC group). Randomisation was done centrally by minimisation with a masked web-based allocation procedure at the time of surgery when residual disease smaller than 10 mm diameter was anticipated, and was stratified by institution, previous suboptimal cytoreductive surgery, and number of abdominal regions involved. The primary endpoint was progression-free survival and a secondary endpoint was overall survival, analysed in the intention-to-treat population (ie, all randomly assigned patients). This study is registered with ClinicalTrials.gov, NCT00426257, and is closed. FINDINGS Between April 1, 2007, and April 30, 2016, 245 patients were enrolled and followed up for a median of 10·1 years (95% CI 8·4-12·9) in the surgery group (n=123) and 10·4 years (95% CI 9·5-13·3) in the surgery-plus-HIPEC group (n=122). Recurrence, progression, or death occurred in 114 (93%) patients in the surgery group (median progression-free survival 10·7 months [95% CI 9·6-12·0]) and 109 (89%) patients in the surgery-plus-HIPEC group (14·3 months [12·0-18·5]; hazard ratio [HR] 0·63 [95% CI 0·48-0·83], stratified log-rank p=0·0008). Death occurred in 108 (88%) patients in the surgery group (median overall survival 33·3 months [95% CI 29·0-39·1]) and 100 (82%) patients in the surgery-plus-HIPEC group (44·9 months [95% CI 38·6-55·1]; HR 0·70 [95% CI 0·53-0·92], stratified log-rank p=0·011). INTERPRETATION These updated survival results confirm the long-term survival benefit of HIPEC in patients with primary stage III epithelial ovarian cancer undergoing interval cytoreductive surgery. FUNDING Dutch Cancer Foundation (KWF Kankerbestrijding).
Collapse
Affiliation(s)
- S Lot Aronson
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands; Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Marta Lopez-Yurda
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Simone N Koole
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands; Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | - Ralph H M Hermans
- Department of Gynecology and Obstetrics, Catharina Hospital, Eindhoven, Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands; Department of Epidemiology, GROW-School for Oncology Reproduction, Maastricht University, Maastricht, Netherlands
| | - Mignon D J M van Gent
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center, Amsterdam, Netherlands; Center for Gynecologic Oncology Amsterdam, Amsterdam, Netherlands
| | - Henriëtte J G Arts
- Department of Gynecologic Oncology, University Medical Center Groningen, Groningen, Netherlands
| | - Maaike A P C van Ham
- Department of Gynecologic Oncology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Peter A van Dam
- Department of Gynecologic Oncology, University Hospital Antwerp, Antwerp, Belgium
| | - Peter Vuylsteke
- Department of Medical Oncology, UCL Louvain, CHU Namur Sainte-Elisabeth, Namur, Belgium; Department of Internal Medicine, University of Botswana, Gaborone, Botswana
| | - Arend G J Aalbers
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | - Neil K Aaronson
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Willemien J van Driel
- Department of Gynecologic Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands; Center for Gynecologic Oncology Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
24
|
Said SA, de Hullu JA, van der Aa MA, Walraven JEW, Bekkers RLM, Slangen BFM, Pickkers P, van Altena AM. Impact of Sepsis on the Oncologic Outcomes of Advanced Epithelial Ovarian Cancer Patients: A Multicenter Observational Study. Cancers (Basel) 2023; 15:4642. [PMID: 37760610 PMCID: PMC10526225 DOI: 10.3390/cancers15184642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/07/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
OBJECTIVE The sepsis-induced inflammatory response may potentially affect malignant cells. Recently, a case of spontaneous regression of a histologically confirmed International Federation of Gynecology and Obstetrics (FIGO) stage IIIC epithelial ovarian cancer (EOC) following sepsis was reported. The aim of our study was to assess the impact of sepsis on the oncologic outcomes of advanced-stage EOC patients. METHODS Gynecologic oncologic patients admitted to the Intensive Care Unit of three oncologic centers between 2006 and 2019 were identified and patients who experienced sepsis following advanced-stage EOC diagnosis were selected. Survival outcomes were compared with advanced-stage EOC patients from the Netherlands Cancer Registry (NCR). To correct for case-mix differences, propensity score matching using 1:3 nearest neighbor matching was conducted after which survival analyses were repeated. RESULTS A total of 18 of 215 patients with advanced-stage EOC experienced sepsis. Sepsis patients had similar distributions of patient, tumor, and treatment characteristics to 3988 patients from the NCR cohort. A total of 3 of 18 patients died from the complications of sepsis. While the remaining patients initially responded to treatment, 14/15 patients relapsed. The median (IQR) overall survival was 31 (24-44) and 35 (20-60) months for the sepsis and unmatched NCR cohort (p = 0.56), respectively. The median (IQR) progression-free survival was 16 (11-21) and 16 (11-27) months (p = 0.90), respectively. Survival outcomes did not differ following propensity matching (overall survival of 31 (24-44) vs. 36 (20-56) months, p = 0.40; progression-free survival of 16 (11-21) and 16 (12-21) months, p = 0.72). CONCLUSION In this observational study, the occurrence of sepsis did not affect the oncologic and survival outcomes of advanced-stage EOC patients.
Collapse
Affiliation(s)
- Sherin A. Said
- Department of Obstetrics and Gynecology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands; (J.A.d.H.); (A.M.v.A.)
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), 3511 DT Utrecht, The Netherlands;
| | - Joanne A. de Hullu
- Department of Obstetrics and Gynecology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands; (J.A.d.H.); (A.M.v.A.)
| | - Maaike A. van der Aa
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), 3511 DT Utrecht, The Netherlands;
| | - Janneke E. W. Walraven
- Department of Medical Oncology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands
| | - Ruud L. M. Bekkers
- Department of Obstetrics and Gynecology, Catharina Hospital, 5623 EJ Eindhoven, The Netherlands
- GROW–School for Oncology and Reproduction, University of Maastricht, 6229 GT Maastricht, The Netherlands
| | - Brigitte F. M. Slangen
- GROW–School for Oncology and Reproduction, University of Maastricht, 6229 GT Maastricht, The Netherlands
- Department of Obstetrics and Gynecology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands;
| | - Anne M. van Altena
- Department of Obstetrics and Gynecology, Radboud University Medical Center, 6525 EP Nijmegen, The Netherlands; (J.A.d.H.); (A.M.v.A.)
| |
Collapse
|
25
|
Bing X, Lei H, Zhao X, Cheng Y, Wang L, Yang J, Xu M, Yu C, Chen T. Use of Period Analysis to Timely Assess Five-Year Relative Survival for Patients with Ovarian Cancer from Taizhou, Eastern China. J Clin Med 2023; 12:jcm12103480. [PMID: 37240586 DOI: 10.3390/jcm12103480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 05/28/2023] Open
Abstract
OBJECTIVE Ovarian cancer is a deadly gynecologic malignancy with a poor prognosis. It is essential to evaluate the early detection and screening programs of ovarian cancer via timely assessment of long-time survival, particularly in China where those data are incredibly limited. Here, we aimed to provide timely and accurately assessment of long-term survival estimate of ovarian cancer patients from eastern China. METHODS Data of 770 ovarian cancer patients diagnosed between 2004-2018 were obtained from four cancer registries in Taizhou, eastern China, were included. We used period analysis to calculate five-year relative survival (RS) of aforementioned ovarian cancer patients for overall and the stratification by age at diagnosis and region. RESULTS Our findings demonstrated that the overall five-year RS for ovarian cancer patients in Taizhou between 2014 and 2018 was 69.2%, while urban areas were higher compared to rural areas (77.6% vs. 64.9%). We also observed a significant age gradient with the five-year RS decreasing from 79.6% for age group < 55 years to 66.9% for age group > 74 years. Furthermore, we identified a clear upward trend of five-year RS over the study period, both overall and stratified by region and age at diagnosis. CONCLUSION This is the first study in China using period analysis to provide the most up-to-date five-year RS for ovarian cancer patients from Taizhou, eastern China, which reaches 69.2% during 2014-2018. Our results provide valuable information for timely assessment of early detection and screening programs for ovarian cancer in eastern China.
Collapse
Affiliation(s)
- Xin Bing
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China
- School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Huijun Lei
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Xiaojiao Zhao
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China
- School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Yongran Cheng
- School of Public Health, Hangzhou Medical College, Hangzhou 310013, China
| | - Liangyou Wang
- Department of Non-Communicable Chronic Disease Control and Prevention, Taizhou Center for Disease Control and Prevention, Taizhou 318000, China
| | - Jun Yang
- School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Mingzhi Xu
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
- Department of General Medicine, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Chenhuan Yu
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Tianhui Chen
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
- Department of Preventive Medicine, School of Medicine, Ningbo University, Ningbo 315211, China
| |
Collapse
|
26
|
Cabasag CJ, Arnold M, Rutherford M, Ferlay J, Bardot A, Morgan E, Butler J, O'Connell DL, Nelson G, Høgdall C, Schnack T, Gavin A, Elwood M, Hanna L, Gourley C, De P, Saint-Jacques N, Mørch LS, Woods RR, Altman AD, Sykes P, Cohen PA, McNally O, Møller B, Walsh P, Morrison DS, Bray F, Soerjomataram I. Shifting incidence and survival of epithelial ovarian cancer (1995-2014): A SurvMark-2 study. Int J Cancer 2023; 152:1763-1777. [PMID: 36533660 DOI: 10.1002/ijc.34403] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/04/2022] [Accepted: 10/25/2022] [Indexed: 12/23/2022]
Abstract
The aim of the study is to provide a comprehensive assessment of incidence and survival trends of epithelial ovarian cancer (EOC) by histological subtype across seven high income countries (Australia, Canada, Denmark, Ireland, New Zealand, Norway and the United Kingdom). Data on invasive EOC diagnosed in women aged 15 to 99 years during 1995 to 2014 were obtained from 20 cancer registries. Age standardized incidence rates and average annual percentage change were calculated by subtype for all ages and age groups (15-64 and 65-99 years). Net survival (NS) was estimated by subtype, age group and 5-year period using Pohar-Perme estimator. Our findings showed marked increase in serous carcinoma incidence was observed between 1995 and 2014 among women aged 65 to 99 years with average annual increase ranging between 2.2% and 5.8%. We documented a marked decrease in the incidence of adenocarcinoma "not otherwise specified" with estimates ranging between 4.4% and 7.4% in women aged 15 to 64 years and between 2.0% and 3.7% among the older age group. Improved survival, combining all EOC subtypes, was observed for all ages combined over the 20-year study period in all countries with 5-year NS absolute percent change ranging between 5.0 in Canada and 12.6 in Denmark. Several factors such as changes in guidelines and advancement in diagnostic tools may potentially influence the observed shift in histological subtypes and temporal trends. Progress in clinical management and treatment over the past decades potentially plays a role in the observed improvements in EOC survival.
Collapse
Affiliation(s)
| | - Melina Arnold
- International Agency for Research on Cancer, Lyon, France
| | - Mark Rutherford
- International Agency for Research on Cancer, Lyon, France
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Jacques Ferlay
- International Agency for Research on Cancer, Lyon, France
| | - Aude Bardot
- International Agency for Research on Cancer, Lyon, France
| | - Eileen Morgan
- International Agency for Research on Cancer, Lyon, France
- Northern Ireland Cancer Registry, Center for Public Health, Queen's University Belfast, Belfast, Northern Ireland, UK
| | | | - Dianne L O'Connell
- The Daffodil Center, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, New South Wales, Australia
| | - Gregg Nelson
- Tom Baker Cancer Center, University of Calgary, Calgary, Alberta, Canada
| | - Claus Høgdall
- Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tine Schnack
- Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Odense University Hospital, Odense, Denmark
| | - Anna Gavin
- Northern Ireland Cancer Registry, Center for Public Health, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Mark Elwood
- Department of Epidemiology and Biostatistics, School of Population Health, University of Auckland, Auckland, New Zealand
| | | | - Charlie Gourley
- Institute of Genetics and Cancer, Cancer Research UK Edinburgh Center, The University of Edinburgh, Edinburgh, Scotland, UK
| | | | | | - Lina Steinrud Mørch
- The Danish Cancer Society Research Center, Cancer Surveillance and Pharmacoepidemiology, Copenhagen, Denmark
| | - Ryan R Woods
- Cancer Control Research, BC Cancer, Vancouver, British Columbia, Canada
| | - Alon D Altman
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Manitoba, Manitoba, Canada
- CancerCare Manitoba Research Institute, Manitoba, Canada
| | - Peter Sykes
- University of Otago, Christchurch, New Zealand
| | - Paul A Cohen
- Department of Gynaecological Oncology, St John of God Subiaco Hospital, Perth, Western Australia, Australia
- Discipline of Obstetrics and Gynaecology, Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Orla McNally
- Royal Women's Hospital, Melbourne, Australia
- University of Melbourne, Melbourne, Australia
| | | | - Paul Walsh
- National Cancer Registry Ireland, Cork, Ireland
| | | | - Freddie Bray
- International Agency for Research on Cancer, Lyon, France
| | | |
Collapse
|
27
|
van Stein RM, Sikorska K, van der Aa MA, Sonke GS, van Driel WJ, van Gent MDJM, van Ham MAPC, Hermans RHM, de Hingh IHJT, Schreuder HWR, The Dutch OVHIPEC‐1 trial group. Evaluation of external validity of the OVHIPEC-1 trial in a real-world population. Int J Gynaecol Obstet 2022; 161:640-648. [PMID: 36495280 DOI: 10.1002/ijgo.14618] [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: 09/18/2022] [Revised: 10/31/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The OVHIPEC-1 trial (Phase III randomised clinical trial for stage III ovarian carcinoma randomising between interval cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy) showed improved survival when interval cytoreductive surgery (CRS) was combined with hyperthermic intraperitoneal chemotherapy in patients with stage III epithelial ovarian cancer (EOC). The authors compared the control arm of the trial with a real-world population treated in the Netherlands during the same period to explore generalizability of the trial results. METHODS For this nationwide comparative cohort study, all patients with EOC undergoing interval CRS between 2007 and 2016 were identified from the Netherlands Cancer Registry if they fulfilled the eligibility criteria of OVHIPEC-1 (n = 1376). Patient and treatment characteristics, and overall survival (OS) were compared between trial and real-world populations. RESULTS Age, comorbidity, BRCA status, histologic subtype, and residual disease were similar in trial and real-world patients. Trial patients had a better performance status, higher socioeconomic status, and underwent bowel surgery more often. In a real-world setting, patients more often received more than six cycles. The difference in OS between the trial and the real-world populations was not statistically significant (unadjusted hazard ratio, 1.09 [95% confidence interval, 0.87-1.37]; P = 0.44). CONCLUSION Despite differences in patient characteristics, OS of patients treated in the control arm of OVHIPEC-1 was similar to patients treated outside the trial. The trial population accurately represents real-world patients with stage III EOC undergoing interval CRS in terms of outcome.
Collapse
Affiliation(s)
- Ruby M van Stein
- Department of Gynecologic Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Karolina Sikorska
- Department of Biometrics, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Maaike A van der Aa
- Department of Research & Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, The Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Willemien J van Driel
- Department of Gynecologic Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Oda K, Aoki D, Tsuda H, Nishihara H, Aoyama H, Inomata H, Shimada M, Enomoto T. Japanese nationwide observational multicenter study of tumor BRCA1/2 variant testing in advanced ovarian cancer. Cancer Sci 2022; 114:271-280. [PMID: 36254756 PMCID: PMC9807512 DOI: 10.1111/cas.15518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 01/07/2023] Open
Abstract
The association between germline BRCA1 and BRCA2 pathogenic variants (mutations: gBRCAm) and ovarian cancer risk is well established. Germline testing alone cannot detect somatic BRCA1/2 pathogenic variants (sBRCAm), which is calculated based on the proportion of tumor BRCAm (tBRCAm) from tumor samples and gBRCAm. Homologous recombination deficiency (HRD) results mainly from genetic/epigenetic alterations in homologous recombination repair-related genes and can be evaluated by genomic instability status. In Japan, the prevalence of tBRCAm, sBRCAm, and HRD remains unclear. This multicenter, cross-sectional, observational study, CHaRacterIzing the croSs-secTional approach to invEstigate the prevaLence of tissue BRCA1/2 mutations in newLy diagnosEd advanced ovarian cancer patients (CHRISTELLE), evaluated the prevalence of tBRCAm, sBRCAm, and HRD in tumor specimens from newly diagnosed patients with ovarian cancer who underwent gBRCA testing. Of the 205 patients analyzed, 26.8% had a tBRCAm, including tBRCA1m (17.6%) and tBRCA2m (9.3%). The overall prevalence of tBRCAm, gBRCAm, sBRCAm, and HRD-positive status was 26.8%, 21.5%, 6.3%, and 60.0%, respectively. The calculated sBRCAm/tBRCAm ratio was 23.6% (13/55), and the prevalence of gBRCA variant of uncertain significance was 3.9%. These results suggest gBRCA testing alone cannot clearly identify the best course of treatment, highlighting the importance of sBRCA testing in Japan. The present results also suggest that testing for tBRCA and HRD should be encouraged in advanced ovarian cancer patients to drive precision medicine.
Collapse
Affiliation(s)
- Katsutoshi Oda
- Division of Integrative Genomics, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Daisuke Aoki
- Department of Obstetrics and GynecologyKeio University School of MedicineTokyoJapan
| | - Hitoshi Tsuda
- Department of Basic PathologyNational Defense Medical CollegeSaitamaJapan
| | | | | | | | - Muneaki Shimada
- Department of Obstetrics and GynecologyTohoku University Graduate School of MedicineMiyagiJapan
| | - Takayuki Enomoto
- Department of Obstetrics and GynecologyNiigata University Medical SchoolNiigataJapan
| |
Collapse
|
29
|
Stur E, Bayraktar E, Dal Molin GZ, Wu SY, Mangala LS, Yao H, Wang Y, Ram PT, Corvigno S, Chen H, Liang H, Tworoger SS, Levine DA, Lutgendorf SK, Liu J, Moore KN, Baggerly KA, Karlan BY, Sood AK. Molecular Analysis of Short- versus Long-Term Survivors of High-Grade Serous Ovarian Carcinoma. Cancers (Basel) 2022; 14:4198. [PMID: 36077735 PMCID: PMC9454595 DOI: 10.3390/cancers14174198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
Despite having similar histologic features, patients with high-grade serous ovarian carcinoma (HGSC) often experience highly variable outcomes. The underlying determinants for long-term survival (LTS, ≥10 years) versus short-term survival (STS, <3 years) are largely unknown. The present study sought to identify molecular predictors of LTS for women with HGSC. A cohort of 24 frozen HGSC samples was collected (12 LTS and 12 STS) and analyzed at DNA, RNA, and protein levels. OVCAR5 and OVCAR8 cell lines were used for in vitro validation studies. For in vivo studies, we injected OVCAR8 cells into the peritoneal cavity of female athymic nude mice. From RNAseq analysis, 11 genes were found to be differentially expressed between the STS and LTS groups (fold change > 2; false discovery rate < 0.01). In the subsequent validation cohort, transmembrane protein 62 (TMEM62) was found to be related to LTS. CIBERSORT analysis showed that T cells (follicular helper) were found at higher levels in tumors from LTS than STS groups. In vitro data using OVCAR5 and OVCAR8 cells showed decreased proliferation with TMEM62 overexpression and positive correlation with a longevity-regulating pathway (KEGG HSA04213) at the RNA level. In vivo analysis using the OVCAR8-TMEM62-TetON model showed decreased tumor burden in mice with high- vs. low-expressing TMEM62 tumors. Our results demonstrate that restoring TMEM62 may be a novel approach for treatment of HGSC. These findings may have implications for biomarker and intervention strategies to help improve patient outcomes
Collapse
Affiliation(s)
- Elaine Stur
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Emine Bayraktar
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Sherry Y. Wu
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Lingegowda S. Mangala
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hui Yao
- Department of Bioinformatics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ying Wang
- Department of Bioinformatics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Prahlad T. Ram
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sara Corvigno
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hu Chen
- Department of Bioinformatics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Han Liang
- Department of Bioinformatics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shelley S. Tworoger
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Douglas A. Levine
- Division of Gynecologic Oncology, New York University, New York, NY 11580, USA
| | - Susan K. Lutgendorf
- Department of Psychological & Brain Sciences, The University of Iowa, Iowa City, IA 52242, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kathleen N. Moore
- Department of Gynecologic Oncology, The University of Oklahoma, Oklahoma City, OK 73117, USA
| | - Keith A. Baggerly
- Department of Bioinformatics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, University of California, Los Angeles, CA 90095, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
30
|
What proportion of patients with stage 3 ovarian cancer are potentially cured following intraperitoneal chemotherapy? Analysis of the long term (≥10 years) survivors in NRG/GOG randomized clinical trials of intraperitoneal and intravenous chemotherapy in stage III ovarian cancer. Gynecol Oncol 2022; 166:410-416. [PMID: 35835612 DOI: 10.1016/j.ygyno.2022.07.004] [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: 03/28/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Patients with advanced epithelial ovarian cancer (EOC) alive without progression at a landmark time-point of 10 years from diagnosis are likely cured. We report the proportion of patients with Stage III EOC who were long-term disease-free survivors (LTDFS≥10 years) following either intraperitoneal (IP) or intravenous (IV) chemotherapy as well as the predictors of LTDFS. METHODS Data from 3 mature NRG/GOG trials (104, 114, 172) were analyzed and included demographics, clinicopathologic details, route of administration, and survival outcomes of patients living ≥10 years assessed according to the Kaplan-Meier method. Cox regression survival analysis was performed to evaluate independent prognostic predictors of LTDFS. RESULTS Of 1174 patients randomized, 10-year overall survival (OS) was 26% (95% CI, 23-28%) and LTDFS ≥10 years was 18% (95% CI, 16-20%). Patients with LTDFS ≥10 years had a median age of 54.6 years (p < 0.001). Younger age (p < 0.001) was the only independent prognostic factor for LTDFS≥10 years on multivariate Cox analysis. CONCLUSIONS Approximately 18% of patients were LTDFS ≥10 years. They form the tail end of the survival curve and are likely cured. Our results provide a comparative benchmark to evaluate the impact of PARP inhibitors in 1st line maintenance trials on survival outcomes.
Collapse
|
31
|
Hassan AA, Artemenko M, Tang MKS, Shi Z, Chen LY, Lai HC, Yang Z, Shum HC, Wong AST. Ascitic fluid shear stress in concert with hepatocyte growth factor drive stemness and chemoresistance of ovarian cancer cells via the c-Met-PI3K/Akt-miR-199a-3p signaling pathway. Cell Death Dis 2022; 13:537. [PMID: 35676254 PMCID: PMC9177676 DOI: 10.1038/s41419-022-04976-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 01/21/2023]
Abstract
Overcoming drug resistance is an inevitable challenge to the success of cancer treatment. Recently, in ovarian cancer, a highly chemoresistant tumor, we demonstrated an important role of shear stress in stem-like phenotype and chemoresistance using a three-dimensional microfluidic device, which most closely mimics tumor behavior. Here, we examined a new mechanosensitive microRNA-miR-199a-3p. Unlike most key microRNA biogenesis in static conditions, we found that Dicer, Drosha, and Exportin 5 were not involved in regulating miR-199a-3p under ascitic fluid shear stress (0.02 dynes/cm2). We further showed that hepatocyte growth factor (HGF), but not other ascitic cytokines/growth factors such as epidermal growth factor and tumor necrosis factor α or hypoxia, could transcriptionally downregulate miR-199a-3p through its primary transcript miR-199a-1 and not miR-199a-2. Shear stress in the presence of HGF resulted in a concerted effect via a specific c-Met/PI3K/Akt signaling axis through a positive feedback loop, thereby driving cancer stemness and drug resistance. We also showed that miR-199a-3p expression was inversely correlated with enhanced drug resistance properties in chemoresistant ovarian cancer lines. Patients with low miR-199a-3p expression were more resistant to platinum with a significantly poor prognosis. miR-199a-3p mimic significantly suppressed ovarian tumor metastasis and its co-targeting in combination with cisplatin or paclitaxel further decreased the peritoneal dissemination of ovarian cancer in mice. These findings unravel how biophysical and biochemical cues regulate miR-199a-3p and is important in chemoresistance. miR-199a-3p mimics may serve as a novel targeted therapy for effective chemosensitization.
Collapse
Affiliation(s)
- Ayon A. Hassan
- grid.194645.b0000000121742757School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong
| | - Margarita Artemenko
- grid.194645.b0000000121742757School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong
| | - Maggie K. S. Tang
- grid.194645.b0000000121742757School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong ,grid.493736.cLaboratory for Synthetic Chemistry and Chemical Biology Limited, 17 W, Hong Kong Science and Technology Parks, Shatin, New Territories Hong Kong
| | - Zeyu Shi
- grid.194645.b0000000121742757School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong
| | - Lin-Yu Chen
- grid.412896.00000 0000 9337 0481Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, Taipei, 23561 Taiwan
| | - Hung-Cheng Lai
- grid.412896.00000 0000 9337 0481Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, Taipei, 23561 Taiwan ,grid.412896.00000 0000 9337 0481Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Zhenyu Yang
- grid.194645.b0000000121742757Department of Mechanical Engineering, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong
| | - Ho-Cheung Shum
- grid.194645.b0000000121742757Department of Mechanical Engineering, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong ,Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories Hong Kong
| | - Alice S. T. Wong
- grid.194645.b0000000121742757School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong
| |
Collapse
|
32
|
Achimas-Cadariu P, Kubelac P, Irimie A, Berindan-Neagoe I, Rühli F. Evolutionary perspectives, heterogeneity and ovarian cancer: a complicated tale from past to present. J Ovarian Res 2022; 15:67. [PMID: 35659345 PMCID: PMC9164402 DOI: 10.1186/s13048-022-01004-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 05/24/2022] [Indexed: 11/21/2022] Open
Abstract
Ovarian cancer is composed of a complex system of cells best described by features such as clonal evolution, spatial and temporal genetic heterogeneity, and development of drug resistance, thus making it the most lethal gynecologic cancer. Seminal work on cancer as an evolutionary process has a long history; however, recent cost-effective large-scale molecular profiling has started to provide novel insights coupled with the development of mathematical algorithms. In the current review, we have systematically searched for articles that focused on the clonal evolution of ovarian cancer to offer the whole landscape of research that has been done and highlight future research avenues given its characteristic features and connections to evolutionary biology.
Collapse
Affiliation(s)
- Patriciu Achimas-Cadariu
- Department of Surgery, The Oncology Institute 'Prof. Dr. Ion Chiricuta', 34-36 Republicii street, 400015 , Cluj-Napoca, Romania. .,Department of Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | - Paul Kubelac
- Department of Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Medical Oncology, The Oncology Institute 'Prof. Dr. Ion Chiricuta', Cluj-Napoca, Romania
| | - Alexandru Irimie
- Department of Surgery, The Oncology Institute 'Prof. Dr. Ion Chiricuta', 34-36 Republicii street, 400015 , Cluj-Napoca, Romania.,Department of Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Centre for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Research Center for Advanced Medicine Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Functional Genomics and Experimental Pathology, The Oncology Institute 'Prof. Dr. Ion Chiricuta', Cluj-Napoca, Romania
| | - Frank Rühli
- Institute of Evolutionary Medicine, Zurich, Switzerland
| |
Collapse
|
33
|
Koole SN, Schouten PC, Hauke J, Kluin RJC, Nederlof P, Richters LK, Krebsbach G, Sikorska K, Alkemade M, Opdam M, Schagen van Leeuwen JH, Schreuder HWR, Hermans RHM, de Hingh IHJT, Mom CH, Arts HJG, van Ham M, van Dam P, Vuylsteke P, Sanders J, Horlings HM, van de Vijver KK, Hahnen E, van Driel WJ, Schmutzler R, Sonke GS, Linn SC. Effect of HIPEC according to HRD/BRCAwt genomic profile in stage III ovarian cancer - results from the phase III OVHIPEC trial. Int J Cancer 2022; 151:1394-1404. [PMID: 35583992 DOI: 10.1002/ijc.34124] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/09/2022] [Accepted: 04/21/2022] [Indexed: 11/07/2022]
Abstract
The addition of hyperthermic intraperitoneal chemotherapy (HIPEC) with cisplatin to interval cytoreductive surgery improves recurrence-free (RFS) and overall survival (OS) in patients with stage III ovarian cancer. Homologous recombination deficient (HRD) ovarian tumors are usually more platinum sensitive. Since hyperthermia impairs BRCA1/2 protein function, we hypothesized that HRD tumors respond best to treatment with HIPEC. We analyzed the effect of HIPEC in patients in the OVHIPEC trial, stratified by HRD status and BRCAm status. Clinical data and tissue samples were collected from patients included in the randomized, phase III OVHIPEC-1 trial. DNA copy number variation (CNV) profiles, HRD-related pathogenic mutations, and BRCA1 promotor hypermethylation were determined. CNV-profiles were categorized as HRD or non-HRD, based on a previously validated algorithm-based BRCA1-like classifier. Hazard ratios (HR) and corresponding 99% confidence intervals (CI) for the effect of RFS and OS of HIPEC in the BRCAm, the HRD/BRCAwt and the non-HRD group were estimated using Cox proportional hazard models. DNA was available from 200/245 (82%) patients. Seventeen (9%) tumors carried a pathogenic mutation in BRCA1 and 14 (7%) in BRCA2. Ninety-one (46%) tumors classified as BRCA1-like. The effect of HIPEC on RFS and OS was absent in BRCAm tumors (HR 1.25; 99%CI 0.48-3.29), and most present in HRD/BRCAwt (HR 0.44; 99%CI 0.21-0.91), and non-HRD/BRCAwt tumors (HR 0.82; 99%CI 0.48-1.42), interaction p-value: 0.024. Patients with HRD tumors without pathogenic BRCA1/2 mutation appear to benefit most from treatment with HIPEC, while benefit in patients with BRCA1/2 pathogenic mutations and patients without HRD seems less evident.
Collapse
Affiliation(s)
- Simone N Koole
- Department of Gynecology, The Netherlands Cancer Institute, Center of Gynecologic Oncology Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Philip C Schouten
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jan Hauke
- Faculty of Medicine and Center for Familial Breast and Ovarian Cancer and Center for Integrated Oncology (CIO), Cologne, University Hospital Cologne, Cologne, Germany
| | - Roel J C Kluin
- Genomics Core Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Petra Nederlof
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lisa K Richters
- Faculty of Medicine and Center for Familial Breast and Ovarian Cancer and Center for Integrated Oncology (CIO), Cologne, University Hospital Cologne, Cologne, Germany
| | - Gabriele Krebsbach
- Faculty of Medicine and Center for Familial Breast and Ovarian Cancer and Center for Integrated Oncology (CIO), Cologne, University Hospital Cologne, Cologne, Germany
| | - Karolina Sikorska
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maartje Alkemade
- Core Facility of Molecular Pathology and Biobanking, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mark Opdam
- Core Facility of Molecular Pathology and Biobanking, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Henk W R Schreuder
- Department of Gynecological Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ralph H M Hermans
- Department of Gynecology and Obstetrics, Catharina Hospital, Eindhoven, The Netherlands
| | | | - Constantijne H Mom
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center, Center of Gynecologic Oncology Amsterdam, Amsterdam, The Netherlands
| | - Henriette J G Arts
- Department of Gynecological Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Maaike van Ham
- Department of Gynecological Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter van Dam
- Department of Gynecologic Oncology, University Hospital Antwerp, Antwerp, Belgium
| | - Peter Vuylsteke
- Department of Medical Oncology, UCL Louvain, CHU Namur Sainte-Elisabeth, Namur, Belgium
- University of Botswana, Gaborone, Botswana
| | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hugo M Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Eric Hahnen
- Faculty of Medicine and Center for Familial Breast and Ovarian Cancer and Center for Integrated Oncology (CIO), Cologne, University Hospital Cologne, Cologne, Germany
| | - Willemien J van Driel
- Department of Gynecology, The Netherlands Cancer Institute, Center of Gynecologic Oncology Amsterdam, Amsterdam, The Netherlands
| | - Rita Schmutzler
- Faculty of Medicine and Center for Familial Breast and Ovarian Cancer and Center for Integrated Oncology (CIO), Cologne, University Hospital Cologne, Cologne, Germany
| | - Gabe S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sabine C Linn
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Sanchez-Cruz P, Vazquez K, Lozada EL, Valiyeva F, Sharma R, Vivas PE, Alegria AE. Photosensitized co-generation of nitric oxide and singlet oxygen Enhanced toxicity against ovarian cancer cells. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2022; 24:82. [PMID: 37035485 PMCID: PMC10081534 DOI: 10.1007/s11051-022-05463-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/23/2022] [Indexed: 06/19/2023]
Abstract
Near micromolar concentrations of nitric oxide (NO) induce tumor cells death. However, an appropriate NO load has to be delivered selectively to the tumor site in order to avoid NO loss and secondary NO-induced effects. The encapsulation of millimolar concentrations of a NO source and an appropriate trigger of NO release within phospatidylcholine-based liposomes should provide an efficient tool for the selective release of the needed NO payload. In this work we report the photosensitized generation of singlet oxygen and NO from folate-targeted PEGylated liposomes, containing AlPcS4 as the sensitizer and S-nitrosoglutathione (GSNO), in millimolar amounts, as the NO source. Amounts of singlet oxygen detected outside the liposome when using PEGylated liposomes are near 200 % larger when GSNO is present inside the liposomes as compared to its absence. These liposomes, conjugated to folate, were found to enhance the photosensitized cytotoxicity to A2780CP20 ovarian cancer cells as compared to liposomes containing the sensitizer but no GSNO (30 % as compared to 70 % cell viability) under the conditions of this work. Fluorescense of AlPcS4 was observed inside cells incubated with folate-conjugated liposomes but not with liposomes without folate. The photosensitized activity enhancement by GSNO increased when light fluence or liposome concentration were increased. The majority of ovarian cancer patients are initially diagnosed with disseminated intra-abdominal disease (stages III-IV) and have a 5-year survival of less than 20%. This work suggests a novel ovarian cancer nodules treatment via the use of tumor-targeted liposome nanoparticles with the capability of generating simultaneously reactive oxygen and nitrogen species upon illumination with near-infrared light.
Collapse
Affiliation(s)
| | - Katerina Vazquez
- Department of Biochemistry, UPR Medical Sciences Campus, San Juan, PR 00936
| | - Eunice L. Lozada
- Comprehensive Cancer Center, UPR Medical Sciences Campus, San Juan, PR 00936
| | - Fatima Valiyeva
- Comprehensive Cancer Center, UPR Medical Sciences Campus, San Juan, PR 00936
| | - Rohit Sharma
- Comprehensive Cancer Center, UPR Medical Sciences Campus, San Juan, PR 00936
| | - Pablo E. Vivas
- Department of Biochemistry, UPR Medical Sciences Campus, San Juan, PR 00936
- Comprehensive Cancer Center, UPR Medical Sciences Campus, San Juan, PR 00936
| | | |
Collapse
|
35
|
Zhu Q(C. A review of co-registered transvaginal Photoacoustic and ultrasound Imaging for Ovarian Cancer Diagnosis. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022; 22. [DOI: 10.1016/j.cobme.2022.100381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
36
|
Liang L, Li J, Yu J, Liu J, Xiu L, Zeng J, Wang T, Li N, Wu L. Establishment and validation of a novel invasion-related gene signature for predicting the prognosis of ovarian cancer. Cancer Cell Int 2022; 22:118. [PMID: 35292033 PMCID: PMC8922755 DOI: 10.1186/s12935-022-02502-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 01/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian cancer (OC) is an invasive gynaecologic cancer with a high cancer-related death rate. The purpose of this study was to establish an invasion-related multigene signature to predict the prognostic risk of OC. METHODS We extracted 97 invasion-related genes from The Cancer Genome Atlas (TCGA) database. Then, the ConsensusClusterPlus and limma packages were used to calculate differentially expressed genes (DEGs). To calculate the immune scores of the molecular subtypes, we used ESTIMATE to evaluate the stromal score, immune score and ESTIMATE score. MCP-counter and the GSVA package ssgsea were used to evaluate the types of infiltrating immune cells. Survival and nomogram analyses were performed to explore the prognostic value of the signature. Finally, qPCR, immunohistochemistry staining and functional assays were used to evaluate the expression and biological abilities of the signature genes in OC. RESULTS Based on the consistent clustering of invasion-related genes, cases in the OC datasets were divided into two subtypes. A significant difference was observed in prognosis between the two subtypes. Most genes were highly expressed in the C1 group. Based on the C1 group genes, we constructed an invasion-related 6-gene prognostic risk model. Furthermore, to verify the signature, we used the TCGA-test and GSE32062 and GSE17260 chip datasets for testing and finally obtained a good risk prediction effect in those datasets. Moreover, the results of the qPCR and immunohistochemistry staining assays revealed that KIF26B, VSIG4 and COL6A6 were upregulated and that FOXJ1, MXRA5 and CXCL9 were downregulated in OC tissues. The functional study showed that the expression of KIF26B, VSIG4, COL6A6, FOXJ1, MXRA5 and CXCL9 can regulate the migration and invasion abilities of OC cells. CONCLUSION We developed a 6-gene prognostic stratification system (FOXJ1, MXRA5, KIF26B, VSIG4, CXCL9 and COL6A6) that is independent of clinical features. These results suggest that the signature could potentially be used to evaluate the prognostic risk of OC patients.
Collapse
Affiliation(s)
- Leilei Liang
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jian Li
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jing Yu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jing Liu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Xiu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jia Zeng
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tiantian Wang
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ning Li
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Lingying Wu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
37
|
de Witte CJ, Kutzera J, van Hoeck A, Nguyen L, Boere IA, Jalving M, Ottevanger PB, van Schaik-van de Mheen C, Stevense M, Kloosterman WP, Zweemer RP, Cuppen E, Witteveen PO. Distinct Genomic Profiles Are Associated with Treatment Response and Survival in Ovarian Cancer. Cancers (Basel) 2022; 14:1511. [PMID: 35326660 PMCID: PMC8946149 DOI: 10.3390/cancers14061511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
The majority of patients with ovarian cancer ultimately develop recurrent chemotherapy-resistant disease. Treatment stratification is mainly based on histological subtype and stage, prior response to platinum-based chemotherapy, and time to recurrent disease. Here, we integrated clinical treatment, treatment response, and survival data with whole-genome sequencing profiles of 132 solid tumor biopsies of metastatic epithelial ovarian cancer to explore genome-informed stratification opportunities. Samples from primary and recurrent disease harbored comparable numbers of single nucleotide variants and structural variants. Mutational signatures represented platinum exposure, homologous recombination deficiency, and aging. Unsupervised hierarchical clustering based on genomic input data identified specific ovarian cancer subgroups, characterized by homologous recombination deficiency, genome stability, and duplications. The clusters exhibited distinct response rates and survival probabilities which could thus potentially be used for genome-informed therapy stratification for more personalized ovarian cancer treatment.
Collapse
Affiliation(s)
- Chris J. de Witte
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (C.J.d.W.); (J.K.); (A.v.H.); (L.N.); (W.P.K.)
| | - Joachim Kutzera
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (C.J.d.W.); (J.K.); (A.v.H.); (L.N.); (W.P.K.)
- Institute of Human Genetics, University Medical Center Leipzig, 04103 Leipzig, Germany
| | - Arne van Hoeck
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (C.J.d.W.); (J.K.); (A.v.H.); (L.N.); (W.P.K.)
| | - Luan Nguyen
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (C.J.d.W.); (J.K.); (A.v.H.); (L.N.); (W.P.K.)
| | - Ingrid A. Boere
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Mathilde Jalving
- Department of Medical Oncology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands;
| | - Petronella B. Ottevanger
- Department of Medical Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | | | - Marion Stevense
- Department of Medical Oncology, Amphia Hospital, 4818 CK Breda, The Netherlands;
| | - Wigard P. Kloosterman
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (C.J.d.W.); (J.K.); (A.v.H.); (L.N.); (W.P.K.)
| | - Ronald P. Zweemer
- Department of Gynaecological Oncology, Cancer Center, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
| | - Edwin Cuppen
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands; (C.J.d.W.); (J.K.); (A.v.H.); (L.N.); (W.P.K.)
- Hartwig Medical Foundation, 1098 XH Amsterdam, The Netherlands
| | - Petronella O. Witteveen
- Department of Medical Oncology, Cancer Center, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
38
|
CaMKK2 Promotes the Progression of Ovarian Carcinoma through the PI3K/PDK1/Akt Axis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7187940. [PMID: 35309839 PMCID: PMC8933102 DOI: 10.1155/2022/7187940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 11/18/2022]
Abstract
Objective. To explore the functional role of Calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) in the progression of ovarian carcinoma (OC). Methods. RT-qPCR analysis and western blot were conducted to detect the mRNA and protein expression of CaMKK2, PI3K, PDK1 and Akt in OC tissues and cells, respectively. CCK-8 assay, transwell migration assay and flow cytometry were used to measure cell proliferation, migration and apoptosis, respectively. Results. CaMKK2, PI3K, PDK1 and Akt were highly expressed in OC tissues compared with the corresponding controls. CaMKK2 knockdown significantly suppressed the mRNA and protein expression of PI3K, PDK1 and Akt in HO8910 and OV90 cells. Moreover, CaMKK2 knockdown could dramatically repress cell proliferation, migration, and markedly elevate cell apoptosis in HO8910 and OV90 cells. Conclusions. CaMKK2 played a promotion role in OC progression via activating the PI3K/PDK1/Akt axis.
Collapse
|
39
|
Vanderstichele A, Loverix L, Busschaert P, Van Nieuwenhuysen E, Han SN, Concin N, Callewaert T, Olbrecht S, Salihi R, Berteloot P, Neven P, Lambrechts D, Van Gorp T, Vergote I. Randomized CLIO/BGOG-ov10 trial of olaparib monotherapy versus physician's choice chemotherapy in relapsed ovarian cancer. Gynecol Oncol 2022; 165:14-22. [PMID: 35177277 DOI: 10.1016/j.ygyno.2022.01.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Comparison of olaparib (OLA) monotherapy versus chemotherapy in patients with platinum-sensitive (PSOC) or platinum-resistant ovarian cancer (PROC). METHODS Patients with measurable disease and ≥ 1 prior line of chemotherapy (CT) were randomized 2:1 to OLA (300 mg tablets, BID) or physician's choice CT.: for PSOC: Carboplatin-Pegylated-Liposomal-Doxorubicin (PLD) or Carboplatin-Gemcitabine; for PROC: PLD, Topotecan, Paclitaxel or Gemcitabine. RESULTS 160 patients (60 with PSOC and 100 with PROC) were randomized 2:1 to OLA (n = 107) or CT (n = 53). Baseline characteristics were similar between both arms. Overall objective response rate (ORR) for OLA and CT were similar (24.3% (26/107) and 28.3% (15/53), respectively). Clinical benefit rate (≥ 12 weeks) was similar with 54.2% (58/107) and 56.6% (30/53), respectively. In PSOC, ORR was 35.0% (14/40) and 65.0% (13/20) for OLA and CT (p = 0.053); in PROC, ORR was 17.9% (12/67) and 6.1% (2/33) for OLA and CT (p = 0.134). ORR in heavily pretreated PROC (>4 prior lines) was 22.9% (8/35) with OLA versus 0% (0/14) for CT. ORR of 35.7% (5/14) and 13.2% (7/53) was observed in BRCA-mutated and -wildtype PROC cases, respectively. Median PFS in PROC was not significantly different with 2.9 months (95% CI 2.8-5.1 in the OLA group versus 3.8 months (95% CI 3.0-6.4) in the CT group (hazard ratio [HR] 1.11 [95% CI 0.72-1.78]; log-rank p = 0.600). CONCLUSION OLA monotherapy showed overall an equal response rate in relapsed ovarian cancer compared with CT. In PROC, ORR and TFST tended to be higher with OLA than with CT. In heavily pretreated patients (four lines or more) with PROC disease, OLA treatment seemed to be more effective than CT.
Collapse
Affiliation(s)
- Adriaan Vanderstichele
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Liselore Loverix
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Pieter Busschaert
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium; Laboratory for Translational Genetics, Department of Oncology, KU Leuven, Leuven, Belgium; Center for Cancer Biology, VIB, Leuven, Belgium
| | - Els Van Nieuwenhuysen
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sileny N Han
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Nicole Concin
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Tiene Callewaert
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Siel Olbrecht
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Rawand Salihi
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Patrick Berteloot
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Patrick Neven
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Oncology, KU Leuven, Leuven, Belgium; Center for Cancer Biology, VIB, Leuven, Belgium
| | - Toon Van Gorp
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Ignace Vergote
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Department of Gynaecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium; Laboratory of Gynaecological Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
40
|
Oncogenic Events Dictate the Types and Locations of Gynecological Malignancies Originating from Krt8+ Mesothelial and Müllerian-Derived Epithelial Cells. Cancers (Basel) 2022; 14:cancers14030841. [PMID: 35159108 PMCID: PMC8834519 DOI: 10.3390/cancers14030841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Ovarian and uterine cancers are the most common gynecological malignancies in women. The early detection, prevention, and treatment of these gynecological cancers can benefit from a better understanding of how tumor-initiating cells in them are formed from their corresponding target cell populations in the female reproductive system. To study this, we utilized a genetic approach in mice to introduce driver mutations commonly found in these cancers to Keratin 8 positive (K8+) mesothelial and epithelial cells in the ovary, fallopian tube, and uterus. We found that p53-loss appears to preferentially affect K8+ epithelial cells, leading to the development of uterine and ovarian malignancies, whereas PTEN-loss may preferentially affect mesothelial cells, leading to the development of ovarian endometrioid malignancies or adenoma on the fallopian tube surface. Collectively, our data suggest that oncogenic driver mutations may dominantly determine the locations and types of gynecological malignancies developed from K8+ mesothelial and epithelial cells in the female reproductive system. Abstract Ovarian and uterine cancers are the most prevalent types of gynecological malignancies originating from mesothelial and/or Müllerian-derived epithelial cells. Recent genomic studies have identified common mutations in them that affect signaling pathways such as p53, PTEN/PI3K, RAS, and WNT pathways. However, how these mutations and their corresponding deregulated pathways affect gynecological cancer development from their cells-of-origin remains largely elusive. To address this, we performed the intrabursal injection of Cre-expressing adenovirus under the control of Krt8 promoter (Ad-K8-Cre) to mice carrying combinations of various conditional alleles for cancer genes. We found that Ad-K8-Cre specifically targeted mesothelial cells, including ovarian surface epithelial (OSE) cells (mainly the LGR5+ subset of OSE cells) and mesothelial cells lining the fallopian tube (FT) serosa; the injected Ad-K8-Cre also targeted Müllerian-derived epithelial cells, including FT epithelial cells and uterine endometrial epithelial cells. The loss of p53 may preferentially affect Müllerian-derived epithelial cells, leading to the development of uterine and ovarian malignancies, whereas PTEN-loss may preferentially affect mesothelial cells, leading to the development of ovarian endometrioid malignancies (upon KRAS-activation or APC-loss) or adenoma on the FT surface (upon DICER-loss). Overall, our data suggest that different Krt8+ mesothelial and epithelial cell types in the female reproductive system may have different sensitivities toward oncogenic mutations and, as a result, oncogenic events may dominantly determine the locations and types of the gynecological malignancies developed from them.
Collapse
|
41
|
David S, Plante A, Dallaire F, Tremblay JP, Sheehy G, Macdonald E, Forrest L, Daneshmand M, Trudel D, Wilson BC, Hopkins L, Murugkar S, Vanderhyden B, Leblond F. Multispectral label-free Raman spectroscopy can detect ovarian and endometrial cancer with high accuracy. JOURNAL OF BIOPHOTONICS 2022; 15:e202100198. [PMID: 34837331 DOI: 10.1002/jbio.202100198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/05/2021] [Accepted: 11/23/2021] [Indexed: 06/13/2023]
Abstract
Up to 70% of ovarian cancer patients are diagnosed with advanced-stage disease and the degree of cytoreduction is an important survival prognostic factor. The aim of this study was to evaluate if Raman spectroscopy could detect cancer from different organs within the abdominopelvic region, including the ovaries. A Raman spectroscopy probe was used to interrogate specimens from a cohort of nine patients undergoing cytoreductive surgery, including four ovarian cancer patients and three patients with endometrial cancer. A feature-selection algorithm was developed to determine which spectral bands contributed to cancer detection and a machine-learning model was trained. The model could detect cancer using only eight spectral bands. The receiver-operating-characteristic curve had an area-under-the-curve of 0.96, corresponding to an accuracy, a sensitivity and a specificity of 90%, 93% and 88%, respectively. These results provide evidence multispectral Raman spectroscopy could be developed to detect ovarian cancer intraoperatively.
Collapse
Affiliation(s)
- Sandryne David
- Department of Engineering Physics, Polytechnique Montreal, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Arthur Plante
- Department of Engineering Physics, Polytechnique Montreal, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Frédérick Dallaire
- Department of Engineering Physics, Polytechnique Montreal, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | | | - Guillaume Sheehy
- Department of Engineering Physics, Polytechnique Montreal, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Elizabeth Macdonald
- Cancer Therapeutics Program, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Laura Forrest
- Cancer Therapeutics Program, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Manijeh Daneshmand
- Cancer Therapeutics Program, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dominique Trudel
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Brian C Wilson
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Laura Hopkins
- Department of Obstetrics and Gynecology, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Barbara Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Frédéric Leblond
- Department of Engineering Physics, Polytechnique Montreal, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| |
Collapse
|
42
|
Winarto H, Welladatika A, Habiburrahman M, Purwoto G, Kusuma F, Utami TW, Putra AD, Anggraeni T, Nuryanto KH. Overall Survival and Related Factors of Advanced-stage Epithelial Ovarian Cancer Patients Underwent Debulking Surgery in Jakarta, Indonesia: A Single-center Experience. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
AIM: The worrisome prognosis of advanced-stage epithelial ovarian cancer (EOC) needs a new perspective from developing countries. Thus, we attempted to study the 5-year overall survival (OS) of advanced-stage EOC patients who underwent debulking surgery in an Indonesian tertiary hospital.
METHODS: A retrospective study recruited forty-eight subjects between 2013 and 2015. We conducted multiple logistic regression analyses to predict risk factors leading to unwanted disease outcomes. The OS was evaluated through the Kaplan–Meier curve and Log-rank test. Cox proportional hazards regression examined prognostic factors of patients.
RESULTS: Prominent characteristics of our patients were middle age (mean: 51.9 ± 8.9 years), obese, with normal menarche onset, multiparous, not using contraception, premenopausal, with serous EOC, and FIGO stage IIIC. The subjects mainly underwent primary debulking surgery (66.8%), with 47.9% of all individuals acquiring optimal results, 77.1% of patients treated had the residual disease (RD), and 52.1% got adjuvant chemotherapy. The risk factor for serous EOC was menopause (odds ratio [OR] = 4.82). The predictors of suboptimal surgery were serous EOC (OR = 8.25) and FIGO stage IV (OR = 11.13). The different OS and median survival were observed exclusively in RD, making it an independent prognostic factor (hazard ratio = 3.50). 5-year A five year OS and median survival for patients with advanced-stage EOC who underwent debulking surgery was 37.5% and 32 months, respectively. Optimal versus suboptimal debulking surgery yielded OS 43.5% versus 32% and median survival of 39 versus 29 months. Both optimal and suboptimal debulking surgery followed with chemotherapy demonstrated an OS 40% lower than those not administered (46.2% and 20%, respectively). The highest 5-year OS was in serous EOC (50%). Meanwhile, the most extended median survival was with mucinous EOC (45 months).
CONCLUSION: Chemotherapy following optimal and suboptimal debulking surgery has the best OS among approaches researched in this study. RD is a significant prognostic factor among advanced-stage EOC. Suboptimal surgery outcomes can be predicted by stage and histological subtype.
Collapse
|
43
|
Ekmann-Gade AW, Høgdall CK, Seibæk L, Noer MC, Fagö-Olsen CL, Schnack TH. Incidence, treatment, and survival trends in older versus younger women with epithelial ovarian cancer from 2005 to 2018: A nationwide Danish study. Gynecol Oncol 2022; 164:120-128. [PMID: 34716025 DOI: 10.1016/j.ygyno.2021.10.081] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To examine clinical trends in Denmark for younger and older epithelial ovarian cancer (EOC) patients, focusing on incidence, treatment, and survival changes. METHODS We included a nationwide cohort diagnosed with EOC from 2005 to 2018. We described age-standardized incidence, surgical patterns, residual disease trends, and cancer-specific survival stratified by age (<70 and ≥ 70 years), stage, and period (2005-09, 2010-13, 2014-18). RESULTS We included 7522 patients. The incidence decreased from 16.3 (2005) to 11.4 (2018) per 100,000 woman-years, driven by the younger cohort. While the proportion of patients with stage IIIC-IV disease undergoing primary debulking surgery (PDS) decreased, the proportion of patients having interval debulking surgery (IDS) and no debulking surgery increased significantly. In 2014-18, 36% and 24% had PDS for younger and older patients, respectively, compared to 72% and 62% in 2005-09. In both age cohorts, the proportion of patients debulked to no residual disease increased significantly among patients with stage IIIC-IV and in the total cohort. Two-year cancer-specific survival increased from 75% (2005-09) to 84% (2014-18) for younger patients and from 53% to 66% for older patients. After adjusting for potential confounders, age ≥ 70 was associated with a 1.4-fold increased risk of cancer-specific death (95% confidence interval: 1.2,1.5). CONCLUSIONS The proportion of patients with advanced EOC not undergoing PDS or IDS increased significantly. During the same period, patients debulked to no residual disease, and cancer-specific survival increased. However, a survival gap in favor of the younger patients remains after adjusting for potential confounders.
Collapse
Affiliation(s)
| | | | - Lene Seibæk
- Department of Gynecology, Aarhus University Hospital, Aarhus, Denmark
| | - Mette Calundann Noer
- Department of Gynecology and Obstetrics, Nordsjællands Hospital, Hillerød, Denmark
| | | | - Tine Henrichsen Schnack
- Department of Gynecology, Rigshospitalet, Copenhagen, Denmark; Department of Gynecology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
44
|
tRNA GlyGCC-Derived Internal Fragment (i-tRF-GlyGCC) in Ovarian Cancer Treatment Outcome and Progression. Cancers (Basel) 2021; 14:cancers14010024. [PMID: 35008188 PMCID: PMC8750938 DOI: 10.3390/cancers14010024] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/21/2022] Open
Abstract
Epithelial ovarian cancer (EOC) remains a highly-lethal gynecological malignancy, characterized by frequent recurrence, chemotherapy resistance and poor 5-year survival. Identifying novel predictive molecular markers remains an overdue challenge in the disease's clinical management. Herein, in silico analysis of TCGA-OV highlighted the tRNA-derived internal fragment (i-tRF-GlyGCC) among the most abundant tRFs in ovarian tumors, while target prediction and gene ontology (GO) enrichment analysis predicted its implication in key biological processes. Thereafter, i-tRF-GlyGCC levels were quantified in a screening EOC (n = 98) and an institutionally-independent serous ovarian cancer (SOC) validation cohort (n = 100, OVCAD multicenter study). Disease progression and patient death were used as clinical endpoints for the survival analysis. Internal validation was performed by bootstrap analysis and the clinical net benefit was estimated by decision curve analysis. The analysis highlighted the significant association of i-tRF-GlyGCC with advanced FIGO stages, suboptimal debulking and most importantly, with early progression and poor overall survival of EOC patients. The OVCAD validation cohort corroborated the unfavorable predictive value of i-tRF-GlyGCC in EOC. Ultimately, evaluation of i-tRF-GlyGCC with the established/clinically used prognostic markers offered superior patient risk-stratification and enhanced clinical benefit in EOC prognosis. In conclusion, i-tRF-GlyGCC assessment could aid towards personalized prognosis and support precision medicine decisions in EOC.
Collapse
|
45
|
Yao T, Rudak PT, Laumont CM, Michaud AR, Rashu R, Knier NN, Foster PJ, McWilliam HEG, Villadangos JA, Nelson BH, DiMattia GE, Shepherd TG, Haeryfar SMM. MAIT cells accumulate in ovarian cancer-elicited ascites where they retain their capacity to respond to MR1 ligands and cytokine cues. Cancer Immunol Immunother 2021; 71:1259-1273. [PMID: 34854949 DOI: 10.1007/s00262-021-03118-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/20/2021] [Indexed: 12/13/2022]
Abstract
The low mutational burden of epithelial ovarian cancer (EOC) is an impediment to immunotherapies that rely on conventional MHC-restricted, neoantigen-reactive T lymphocytes. Mucosa-associated invariant T (MAIT) cells are MR1-restricted T cells with remarkable immunomodulatory properties. We sought to characterize intratumoral and ascitic MAIT cells in EOC. Single-cell RNA sequencing of six primary human tumor specimens demonstrated that MAIT cells were present at low frequencies within several tumors. When detectable, these cells highly expressed CD69 and VSIR, but otherwise exhibited a transcriptomic signature inconsistent with overt cellular activation and/or exhaustion. Unlike mainstream CD8+ T cells, CD8+ MAIT cells harbored high transcript levels of TNF, PRF1, GZMM and GNLY, suggesting their arming and cytotoxic potentials. In a congenic, MAIT cell-sufficient mouse model of EOC, MAIT and invariant natural killer T cells amassed in the peritoneal cavity where they showed robust IL-17A and IFN-γ production capacities, respectively. However, they gradually lost these functions with tumor progression. In a cohort of 23 EOC patients, MAIT cells were readily detectable in all ascitic fluids examined. In a sub-cohort in which we interrogated ascitic MAIT cells for functional impairments, several exhaustion markers, most notably VISTA, were present on the surface. However, ascitic MAIT cells were capable of producing IFN-γ, TNF-α and granzyme B, but neither IL-17A nor IL-10, in response to an MR1 ligand, bacterial lysates containing MR1 ligands, or a combination of IL-12 and IL-18. In conclusion, ascitic MAIT cells in EOC possess inducible effector functions that may be modified in future immunotherapeutic strategies.
Collapse
Affiliation(s)
- Tony Yao
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, Canada
| | - Patrick T Rudak
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, Canada
| | - Céline M Laumont
- Deeley Research Centre, BC Cancer, Victoria, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Alex R Michaud
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, Canada
| | - Rasheduzzaman Rashu
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, Canada
| | - Natasha N Knier
- Department of Medical Biophysics, Western University, London, ON, Canada.,Robarts Research Institute, Western University, London, ON, Canada
| | - Paula J Foster
- Department of Medical Biophysics, Western University, London, ON, Canada.,Robarts Research Institute, Western University, London, ON, Canada
| | - Hamish E G McWilliam
- Department of Microbiology and Immunology, The Peter Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.,Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Jose A Villadangos
- Department of Microbiology and Immunology, The Peter Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.,Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Gabriel E DiMattia
- Department of Biochemistry, Western University, London, ON, Canada.,The Mary & John Knight Translational Ovarian Cancer Research Unit, London, ON, Canada.,Department of Oncology, Western University, London, ON, Canada.,Department of Obstetrics and Gynaecology, Western University, London, ON, Canada.,London Regional Cancer Program, London, ON, Canada.,Lawson Health Research Institute, London, ON, Canada
| | - Trevor G Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London, ON, Canada.,Department of Oncology, Western University, London, ON, Canada.,Department of Obstetrics and Gynaecology, Western University, London, ON, Canada.,London Regional Cancer Program, London, ON, Canada.,Lawson Health Research Institute, London, ON, Canada.,Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, Canada. .,Lawson Health Research Institute, London, ON, Canada. .,Centre for Human Immunology, Western University, London, ON, Canada. .,Department of Surgery, Division of General Surgery, Western University, London, ON, Canada. .,Department of Medicine, Division of Clinical Immunology and Allergy, Western University, London, ON, Canada.
| |
Collapse
|
46
|
Liang Z, Dong J, Yang N, Li SD, Yang ZY, Huang R, Li FJ, Wang WT, Ren JK, Lei J, Xu C, Wang D, Wang YZ, Liang ZQ. Tandem CAR-T cells targeting FOLR1 and MSLN enhance the antitumor effects in ovarian cancer. Int J Biol Sci 2021; 17:4365-4376. [PMID: 34803504 PMCID: PMC8579462 DOI: 10.7150/ijbs.63181] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/30/2021] [Indexed: 11/05/2022] Open
Abstract
Given the heterogeneity of solid tumors, single-target CAR-T cell therapy often leads to recurrence, especially in ovarian cancer (OV). Here, we constructed a Tandem-CAR targeting two antigens with secretory activity (IL-12) to improve the effects of CAR-T cell therapy. Twenty coexpressed upregulated genes were identified from the GEO database, and we found FOLR1 (folate receptor 1) and MSLN (mesothelin) were specifically and highly expressed in cancer tissues and only 11.25% of samples were negative for both antigens. We observed an increased proliferation rate for these three CAR-T cells, and Tandem CAR-T cells could efficiently lyse antigen-positive OV cells in vitro and secrete higher levels of cytokines than single-target CAR-T cells. More importantly, in vivo experiments indicated that Tandem CAR-T cells markedly decreased tumor volume, exhibited enhanced antitumor activity, and prolonged mouse survival. Furthermore, the infiltration and persistence of T cells in the Tandem-CAR group were higher than those in the MSLN-CAR and Control-T groups but comparable to those in the FOLR1-CAR group. Collectively, this study demonstrated that Tandem CAR-T cells secreting IL-12 could enhance immunotherapeutic effects by reducing tumor antigen escape and increasing T cell functionality, which could be a promising therapeutic strategy for OV and other solid tumors.
Collapse
Affiliation(s)
- Zhen Liang
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiao Dong
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Obstetrics and Gynecology, Guangyuan Traditional Chinese Medicine Hospital, Guangyuan, China
| | - Neng Yang
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Si-Di Li
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ze-Yu Yang
- Breast and Thyroid Surgical Department, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Rui Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Feng-Jie Li
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wen-Ting Wang
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jia-Kui Ren
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jie Lei
- Department of Internal Medicine, Hui Long-Ba Town Hospital, Chongqing, China
| | - Chen Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Dan Wang
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan-Zhou Wang
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhi-Qing Liang
- Department of Obstetrics and Gynecology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
47
|
Wenzel L, Osann K, McKinney C, Cella D, Fulci G, Scroggins MJ, Lankes HA, Wang V, Nephew KP, Maxwell GL, Mok SC, Conrads TP, Miller A, Mannel RS, Gray HJ, Hanjani P, Huh WK, Spirtos N, Leitao MM, Glaser G, Sharma SK, Santin AD, Sperduto P, Lele SB, Burger RA, Monk BJ, Birrer M. Quality of Life and Adverse Events: Prognostic Relationships in Long-Term Ovarian Cancer Survival. J Natl Cancer Inst 2021; 113:1369-1378. [PMID: 33729494 PMCID: PMC8486331 DOI: 10.1093/jnci/djab034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/15/2021] [Accepted: 03/05/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND There is a critical need to identify patient characteristics associated with long-term ovarian cancer survival. METHODS Quality of life (QOL), measured by the Functional Assessment of Cancer Therapy-Ovarian-Trial Outcome Index (FACT-O-TOI), including physical, functional, and ovarian-specific subscales, was compared between long-term survivors (LTS) (8+ years) and short-term survivors (STS) (<5 years) of GOG 218 at baseline; before cycles 4, 7, 13, 21; and 6 months post-treatment using linear and longitudinal mixed models adjusted for covariates. Adverse events (AEs) were compared between survivor groups at each assessment using generalized linear models. All P values are 2-sided. RESULTS QOL differed statistically significantly between STS (N = 1115) and LTS (N = 260) (P < .001). Baseline FACT-O-TOI and FACT-O-TOI change were independently associated with long-term survival (odds ratio = 1.05, 95% confidence interval = 1.03 to 1.06 and odds ratio = 1.06, 95% confidence interval = 1.05 to 1.07, respectively). A 7-point increase in baseline QOL was associated with a 38.0% increase in probability of LTS, and a 9-point increase in QOL change was associated with a 67.0% increase in odds for LTS. QOL decreased statistically significantly with increasing AE quartiles (cycle 4 quartiles: 0-5 vs 6-8 vs 9-11 vs ≥12 AEs, P = .01; cycle 21 quartiles: 0-2 vs 3 vs 4-5 vs ≥6 AEs, P = .001). Further, LTS reported statistically significantly better QOL compared with STS (P = .03 and P = .01, cycles 4 and 21, respectively), with similar findings across higher AE grades. CONCLUSIONS Baseline and longitudinal QOL change scores distinguished LTS vs STS and are robust prognosticators for long-term survival. Results have trial design and supportive care implications, providing meaningful prognostic value in this understudied population.
Collapse
Affiliation(s)
- Lari Wenzel
- Department of Medicine and Program in Public Health, University of California, Irvine, CA, USA
| | - Kathryn Osann
- Department of Medicine and Program in Public Health, University of California, Irvine, CA, USA
| | - Chelsea McKinney
- Department of Medicine and Program in Public Health, University of California, Irvine, CA, USA
| | - David Cella
- Department of Medical Social Sciences, Northwestern University Health System, Chicago, IL, USA
| | | | | | | | - Victoria Wang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth P Nephew
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, IN, USA
| | - George L Maxwell
- Women’s Health Integrated Research Center at Inova Health System, Women’s Service Line, Inova Health System, Falls Church, VA, USA
| | - Samuel C Mok
- Department of Gynecological Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas P Conrads
- Women’s Health Integrated Research Center at Inova Health System, Women’s Service Line, Inova Health System, Falls Church, VA, USA
| | - Austin Miller
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Robert S Mannel
- Stephenson Cancer Center, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Oklahoma, Oklahoma City, OK, USA
| | - Heidi J Gray
- Gynecologic Oncology, University of Washington Medical Center, Seattle, WA, USA
| | - Parviz Hanjani
- Gynecologic Oncology, Abington—Jefferson Health, Abington, PA, USA
| | - Warner K Huh
- University of Alabama Highlands, Birmingham, AL, USA
| | - Nick Spirtos
- Women’s Cancer Center of Nevada, Las Vegas, NV, USA
| | - Mario M Leitao
- Memorial Sloan Kettering Cancer and Weill Cornell Medical Center, New York, NY, USA
| | | | | | - Alessandro D Santin
- Department of Obstetrics, Gynecology and Reproductive Services, Yale University School of Medicine, New Haven, CT, USA
| | - Paul Sperduto
- Minneapolis Radiation Oncology and Metro-Minnesota Community Oncology Research Consortium, St. Louis Park, MN, USA
| | | | | | - Bradley J Monk
- Division of Gynecologic Oncology, Arizona Oncology (US Oncology Network), University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Michael Birrer
- Winthrop P. Rockefeller Cancer Institute University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
48
|
A New Paradigm in Managing Advanced Ovarian Cancer: Differentiating Patients Requiring Neoadjuvant Treatment from Primary Cytoreduction. Cancers (Basel) 2021; 13:cancers13194925. [PMID: 34638409 PMCID: PMC8508489 DOI: 10.3390/cancers13194925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 01/22/2023] Open
Abstract
Simple Summary This study evaluates the comparability of patients treated with primary cytoreduction and patients undergoing neoadjuvant chemotherapy for advanced stages high grade serous ovarian carcinoma by comparing the preoperative and postoperative characteristics after a propensity score matching analysis during ten years in a tertiary cancer center. Abstract Our study aims to evaluate the comparability of primary debulking surgery (PDS) and neoadjuvant chemotherapy (NACT) patients. This single-center retrospective study includes all patients treated for advanced stages high-grade serous ovarian carcinomas (HGSOC) between 2007 and 2017. Preoperative characteristics and postoperative outcomes were compared after a propensity score matching analysis. Of the 221 patients included, 38% underwent PDS, and 62% received NACT. There was no age difference at diagnosis; however, CA125 levels, PCI score levels, and rates of stage IV were higher in the NACT group. There were no differences concerning the rate and the severity of complications (p = 0.29). The propensity score distribution showed a broad distinction between PDS patients and NACT patients with no significant overlap. Survival analyses demonstrate, after a median follow-up of 66.5 months, an overall survival (OS) of 105.9 and progression-free survival (PFS) of 29.2 months in the PDS group, compared to OS of 52.8 and PFS of 18.9 months in the NACT group. Advanced HGSOC is a heterogeneous population, in which inoperable patients should be differentiated from PDS patients based on many factors, primarily tumor burden.
Collapse
|
49
|
Engbersen MP, Van Driel W, Lambregts D, Lahaye M. The role of CT, PET-CT, and MRI in ovarian cancer. Br J Radiol 2021; 94:20210117. [PMID: 34415198 DOI: 10.1259/bjr.20210117] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
New treatment developments in ovarian cancer have led to a renewed interest in staging advanced ovarian cancer. The treatment of females with ovarian cancer patients has a strong multidisciplinary character with an essential role for the radiologist. This review aims to provide an overview of the current position of CT, positron emission tomography-CT, and MRI in ovarian cancer and how imaging can be used to guide multidisciplinary team discussions.
Collapse
Affiliation(s)
- Maurits Peter Engbersen
- Department of Radiology, Antoni van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Willemien Van Driel
- Department of Gynecology, Center of Gynecological Oncology Amsterdam, Antoni van Leeuwenhoek- Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Doenja Lambregts
- Department of Radiology, Antoni van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Max Lahaye
- Department of Radiology, Antoni van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
50
|
A HER2 Tri-Specific NK Cell Engager Mediates Efficient Targeting of Human Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13163994. [PMID: 34439149 PMCID: PMC8394622 DOI: 10.3390/cancers13163994] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary HER2 is a marker known to be over-expressed on breast cancer, rendering it one of the most useful solid tumor targets for antibody-based therapies. Despite expression on ovarian cancer, results targeting HER2 in this setting have been disappointing, thus requiring more aggressive approaches. Natural killer (NK) cells are known as principal mediators of cancer cell killing, but cancer cells find ways to deter them. We devised a tri-specific biological drug containing antibody fragments that simultaneously binds NK cells and cancer cells and at the same time delivers a natural cytokine signal that triggers robust NK cell expansion. In vitro studies show the drug augments NK cell killing of a number of HER2-positive human cell lines, while enhancing NK cell activation and proliferation. Studies in mice engrafted with human ovarian cancer showed the drug has anti-tumor efficacy, clearly demonstrating its ability to bolster NK cells in their ability to contain tumor cell growth. Abstract Clinical studies validated antibodies directed against HER2, trastuzumab, and pertuzumab, as useful methodology to target breast cancer cases where HER2 is expressed. The hope was that HER2 targeting using these antibodies in ovarian cancer patients would prove useful as well, but clinical studies have shown lackluster results in this setting, indicating a need for a more comprehensive approach. Immunotherapy approaches stimulating the innate immune system show great promise, although enhancing natural killer (NK) function is not an established mainstream immunotherapy. This study focused on a new nanobody platform technology in which the bispecific antibody was altered to incorporate a cytokine. Herein we describe bioengineered CAM1615HER2 consisting of a camelid VHH antibody fragment recognizing CD16 and a single chain variable fragment (scFv) recognizing HER2 cross-linked by the human interleukin-15 (IL-15) cytokine. This tri-specific killer engager (TriKETM) showed in vitro prowess in its ability to kill ovarian cancer human cell lines. In addition, we demonstrated its efficacy in inducing potent anti-cancer effects in an in vivo xenograft model of human ovarian cancer engrafting both cancer cells and human NK cells. While previous approaches with trastuzumab and pertuzumab faltered in ovarian cancer, the hope is incorporating targeting and cytokine priming within the same molecule will enhance efficacy in this setting.
Collapse
|