1
|
Matboli M, Al-Amodi HS, Khaled A, Khaled R, Ali M, Kamel HFM, Hamid MSAEL, ELsawi HA, Habib EK, Youssef I. Integrating molecular, biochemical, and immunohistochemical features as predictors of hepatocellular carcinoma drug response using machine-learning algorithms. Front Mol Biosci 2024; 11:1430794. [PMID: 39479501 PMCID: PMC11521808 DOI: 10.3389/fmolb.2024.1430794] [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: 05/10/2024] [Accepted: 09/27/2024] [Indexed: 11/02/2024] Open
Abstract
Introduction Liver cancer, particularly Hepatocellular carcinoma (HCC), remains a significant global health concern due to its high prevalence and heterogeneous nature. Despite the existence of approved drugs for HCC treatment, the scarcity of predictive biomarkers limits their effective utilization. Integrating diverse data types to revolutionize drug response prediction, ultimately enabling personalized HCC management. Method In this study, we developed multiple supervised machine learning models to predict treatment response. These models utilized classifiers such as logistic regression (LR), k-nearest neighbors (kNN), neural networks (NN), support vector machines (SVM), and random forests (RF) using a comprehensive set of molecular, biochemical, and immunohistochemical features as targets of three drugs: Pantoprazole, Cyanidin 3-glycoside (Cyan), and Hesperidin. A set of performance metrics for the complete and reduced models were reported including accuracy, precision, recall (sensitivity), specificity, and the Matthews Correlation Coefficient (MCC). Results and Discussion Notably, (NN) achieved the best prediction accuracy where the combined model using molecular and biochemical features exhibited exceptional predictive power, achieving solid accuracy of 0.9693 ∓ 0.0105 and average area under the ROC curve (AUC) of 0.94 ∓ 0.06 coming from three cross-validation iterations. Also, found seven molecular features, seven biochemical features, and one immunohistochemistry feature as promising biomarkers of treatment response. This comprehensive method has the potential to significantly advance personalized HCC therapy by allowing for more precise drug response estimation and assisting in the identification of effective treatment strategies.
Collapse
Affiliation(s)
- Marwa Matboli
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Faculty of Oral and Dental Medicine, Misr International University (MIU), Cairo, Egypt
| | - Hiba S. Al-Amodi
- Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdelrahman Khaled
- Bioinformatics Group, Center of Informatics Sciences (CIS), School of Information Technology and Computer Sciences, Nile University, Giza, Egypt
| | - Radwa Khaled
- Biotechnology/Biomolecular Chemistry Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Marwa Ali
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hala F. M. Kamel
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Hind A. ELsawi
- Department of Internal Medicine, Badr University in Cairo, Badr, Egypt
| | - Eman K. Habib
- Department of Anatomy and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Anatomy and Cell Biology, Faculty of Medicine, Galala University, Suez, Egypt
| | - Ibrahim Youssef
- Systems and Biomedical Engineering Department, Faculty of Engineering, Cairo University, Giza, Egypt
| |
Collapse
|
2
|
Azzalini E, Di Stefano B, Canzonieri V, Venesio T, Miglio U, Marchiò C, Sapino A, Previderè C, Fattorini P, Bonin S. Quantifying mRNA in Highly Degraded Fixed Tissues by Nanostring Technology: A Comparative Study. Methods Protoc 2024; 7:40. [PMID: 38804334 PMCID: PMC11130824 DOI: 10.3390/mps7030040] [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: 03/27/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/29/2024] Open
Abstract
Archive tissues are the most available source of human tissues useful for molecular analysis in translational research. The main issues for those specimens are the modification and degradation of biomolecules, namely proteins, DNA, and RNA. In the last decade, several high-throughput analytical methods have been applied to archive tissues. Although histological tissues are fixed in neutral-buffered formalin nowadays, in the recent past, Bouin's solution was also used in tissue processing. The present study aims to investigate the feasibility of nCounter Nanostring hybridization in quantifying mRNA in highly degraded samples, such as Bouin's fixed and paraffin-embedded (BFPE) tissues, in comparison to the standard formalin-fixed and paraffin-embedded (FFPE) tissues as a source of RNA. A total of 16 paraffin-embedded tissue blocks from eight patients were analyzed (8 were FFPE and 8 were BEPE). Nanostring technology was applied to 300 ng of each RNA sample, whereas 360 ng of the same templates were retrotranscribed and submitted to qPCR and ddPCR. Our results show that the Nanostring technology outperforms the reference methods (ddPCR and qPCR) in detecting target mRNA in FFPE and BFPE samples. However, even Nanostring technology does not escape the limitation imposed by the degradation of the RNA templates, which could lead to misleading conclusions on the gene expression level.
Collapse
Affiliation(s)
- Eros Azzalini
- Department of Medical Sciences, University of Trieste, 34149 Trieste, Italy; (E.A.); (B.D.S.); (V.C.); (S.B.)
| | - Barbara Di Stefano
- Department of Medical Sciences, University of Trieste, 34149 Trieste, Italy; (E.A.); (B.D.S.); (V.C.); (S.B.)
| | - Vincenzo Canzonieri
- Department of Medical Sciences, University of Trieste, 34149 Trieste, Italy; (E.A.); (B.D.S.); (V.C.); (S.B.)
- Pathology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano-National Cancer Institute, 33081 Pordenone, Italy
| | - Tiziana Venesio
- Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-IRCCS, 10060 Candiolo, Italy; (T.V.); (U.M.); (C.M.); (A.S.)
| | - Umberto Miglio
- Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-IRCCS, 10060 Candiolo, Italy; (T.V.); (U.M.); (C.M.); (A.S.)
| | - Caterina Marchiò
- Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-IRCCS, 10060 Candiolo, Italy; (T.V.); (U.M.); (C.M.); (A.S.)
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-IRCCS, 10060 Candiolo, Italy; (T.V.); (U.M.); (C.M.); (A.S.)
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Carlo Previderè
- Laboratorio di Genetica Forense, Dipartimento di Sanità Pubblica, Medicina Sperimentale e Forense, Università di Pavia, 27100 Pavia, Italy;
| | - Paolo Fattorini
- Department of Medical Sciences, University of Trieste, 34149 Trieste, Italy; (E.A.); (B.D.S.); (V.C.); (S.B.)
| | - Serena Bonin
- Department of Medical Sciences, University of Trieste, 34149 Trieste, Italy; (E.A.); (B.D.S.); (V.C.); (S.B.)
| |
Collapse
|
3
|
DasGupta R, Yap A, Yaqing EY, Chia S. Evolution of precision oncology-guided treatment paradigms. WIREs Mech Dis 2023; 15:e1585. [PMID: 36168283 DOI: 10.1002/wsbm.1585] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 01/31/2023]
Abstract
Cancer treatment is gradually evolving from the classical use of nonspecific cytotoxic drugs targeting generic mechanisms of cell growth and proliferation. Instead, new "patient-specific treatment paradigms" that are based on an individual patient's tumor-specific molecular features are emerging, and these include "druggable" genomic alterations such as oncogenic driver mutations, downstream activities of cancer-signaling pathways, and the expression of specific genes involved in tumorigenesis and cancer progression. This evolving landscape of making evidence-based treatment decisions forms the foundation of precision oncology, which aims to deliver "the right drug, to the right patient and at the right time". The long-term vision for this approach is to maximize the treatment efficacy while minimizing exposure to ineffective therapy and reducing co-morbidity-related side effects. Successful clinical translation and implementation of this vision have the potential to revolutionize treatment paradigms from predominantly reactive, to more evidence-based, proactive and predictive care. In this article, we review the past and current approaches in precision oncology, and describe their remarkable power and limitations. We also speculate on the evolution of newly emerging methodologies of the future that can be used to address some of the key challenges associated with the existing paradigms. This article is categorized under: Cancer > Genetics/Genomics/Epigenetics Cancer > Molecular and Cellular Physiology Cancer > Computational Models.
Collapse
Affiliation(s)
- Ramanuj DasGupta
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore.,Cancer Science Institute, National University of Singapore, Singapore, Singapore
| | - Aixin Yap
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Elena Yong Yaqing
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Shumei Chia
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| |
Collapse
|
4
|
Bellcross CA. Hereditary Breast and Ovarian Cancer. Obstet Gynecol Clin North Am 2022; 49:117-147. [DOI: 10.1016/j.ogc.2021.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
5
|
Azzalini E, Barbazza R, Stanta G, Giorda G, Bortot L, Bartoletti M, Puglisi F, Canzonieri V, Bonin S. Histological patterns and intra-tumor heterogeneity as prognostication tools in high grade serous ovarian cancers. Gynecol Oncol 2021; 163:498-505. [PMID: 34602289 DOI: 10.1016/j.ygyno.2021.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 08/24/2021] [Accepted: 09/19/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE High grade serous ovarian carcinoma (HGSOC) is the most common type of malignant ovarian neoplasm and the main cause of ovarian cancer related deaths worldwide. Although novel biomarkers such as homologous recombination deficiency testing have been implemented into the clinical decision-making algorithm since diagnosis, morphological classification and immunohistochemistry analysis are essential for diagnostic purpose. This study aims at identifying histologic and clinical features that can be predictive of patients' prognosis. METHODS Morphological and architectural characterization including SET (Solid-Endometroid-Transitional)/Classic features was carried out in a cohort of 234 patients analyzing 695 slides. From each slide tumor infiltrating lymphocyte (TILs), the presence of necrosis, the number of mitoses, the presence of psammoma bodies, giant cells and atypical mitoses were recorded. Morphological heterogeneity was quantified by the Shannon's diversity index (SDI) considering the percentage of each architectural pattern per patient's slide. RESULTS The frequency of architectural patterns and morphological variables varied with respect of the surgical strategy (primary debulking surgery vs interval surgery after neoadjuvant chemotherapy). HGSOCs exhibiting SET features had a longer overall as well as progression free survival. Among SET features, pseudo-endometrioid and transitional like patterns had the best outcome, while it was heterogenous for solid pattern, that had better outcome for BRCA 1 negative and less heterogeneous tumors. In patients submitted to neoadjuvant chemotherapy a higher intratumor heterogeneity as defined by SDI was a negative independent prognostic factor. CONCLUSIONS A comprehensive histological examination considering architectural patterns and their heterogeneity can help in prognostication of HGSOCs.
Collapse
Affiliation(s)
- Eros Azzalini
- DSM- Department of Medical Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; IRCCS CRO Aviano-National Cancer Institute, Via Gallini 2, 33081 Aviano, Italy
| | - Renzo Barbazza
- DSM- Department of Medical Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Giorgio Stanta
- DSM- Department of Medical Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Giorgio Giorda
- IRCCS CRO Aviano-National Cancer Institute, Via Gallini 2, 33081 Aviano, Italy
| | - Lucia Bortot
- Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, PN, Italy; DAME - Department of Medicine, University of Udine, Via Colugna 50, 33100 Udine, Italy
| | - Michele Bartoletti
- Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, PN, Italy; DAME - Department of Medicine, University of Udine, Via Colugna 50, 33100 Udine, Italy
| | - Fabio Puglisi
- Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, PN, Italy; DAME - Department of Medicine, University of Udine, Via Colugna 50, 33100 Udine, Italy
| | - Vincenzo Canzonieri
- Pathology Unit, IRCCS CRO Aviano-National Cancer Institute, Via Gallini 2, 33081 Aviano, Italy; DSM- Department of Medical Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy.
| | - Serena Bonin
- DSM- Department of Medical Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy.
| |
Collapse
|
6
|
BECN1 and BRCA1 Deficiency Sensitizes Ovarian Cancer to Platinum Therapy and Confers Better Prognosis. Biomedicines 2021; 9:biomedicines9020207. [PMID: 33670664 PMCID: PMC7922320 DOI: 10.3390/biomedicines9020207] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/21/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Background: BRCA1, BECN1 and TP53 are three tumor suppressor genes located on chromosome 17 and frequently found deleted, silenced, or mutated in many cancers. These genes are involved in autophagy, apoptosis, and drug resistance in ovarian cancer. Haploinsufficiency or loss-of-function of either TP53, BRCA1 or BECN1 correlates with enhanced predisposition to cancer development and progression, and chemoresistance. Expectedly, the combined altered expression of these three tumor suppressor genes worsens the prognosis of ovarian cancer patients. However, whether such a genotypic pattern indeed affects the chemo-responsiveness to standard chemotherapy thus worsening patients’ survival has not been validated in a large cohort of ovarian cancer patients. Aim: We interrogated datasets from the TCGA database to analyze how the expression of these three tumor suppressor genes impacts on the clinical response to platinum-based chemotherapy thus affecting the survival of ovarian cancer patients. Results and conclusion: Compared to EOC with homozygous expression of BECN1 and BRCA1, tumors expressing low mRNA expression of these two tumor suppressor genes (either because of shallow (monoallelic) co-deletion or of promoter hypermethylation), showed higher sensitivity to platinum-based therapies and were associated with a better prognosis of ovarian cancer-bearing patients. This outcome was independent of TP53 status, though it was statistically more significant in the cohort of patients with mutated TP53. Thus, sensitivity to platinum therapy (and probably to other chemotherapeutics) correlates with low expression of a combination of critical tumor suppressor genes. Our study highlights the importance of thoroughly assessing the genetic lesions of the most frequently mutated genes to stratify the patients in view of a personalized therapy. More importantly, the present findings suggest that targeting the function of both BECN1 and BRCA1 could be a strategy to restore chemosensitivity in refractory tumors.
Collapse
|
7
|
Li K, Du Y, Li L, Wei DQ. Bioinformatics Approaches for Anti-cancer Drug Discovery. Curr Drug Targets 2021; 21:3-17. [PMID: 31549592 DOI: 10.2174/1389450120666190923162203] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 07/17/2019] [Accepted: 07/26/2019] [Indexed: 12/23/2022]
Abstract
Drug discovery is important in cancer therapy and precision medicines. Traditional approaches of drug discovery are mainly based on in vivo animal experiments and in vitro drug screening, but these methods are usually expensive and laborious. In the last decade, omics data explosion provides an opportunity for computational prediction of anti-cancer drugs, improving the efficiency of drug discovery. High-throughput transcriptome data were widely used in biomarkers' identification and drug prediction by integrating with drug-response data. Moreover, biological network theory and methodology were also successfully applied to the anti-cancer drug discovery, such as studies based on protein-protein interaction network, drug-target network and disease-gene network. In this review, we summarized and discussed the bioinformatics approaches for predicting anti-cancer drugs and drug combinations based on the multi-omic data, including transcriptomics, toxicogenomics, functional genomics and biological network. We believe that the general overview of available databases and current computational methods will be helpful for the development of novel cancer therapy strategies.
Collapse
Affiliation(s)
- Kening Li
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuxin Du
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lu Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing 211166, China
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
8
|
Comandante-Lou N, Fallahi-Sichani M. Models of Cancer Drug Discovery and Response to Therapy. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11356-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
9
|
Li S, Zhu Z. Chemotherapy is not necessary for early-stage serous and endometrioid ovarian cancer after undergoing comprehensive staging surgery. J Ovarian Res 2020; 13:91. [PMID: 32772926 PMCID: PMC7416408 DOI: 10.1186/s13048-020-00694-9] [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: 06/11/2020] [Accepted: 07/28/2020] [Indexed: 11/24/2022] Open
Abstract
In order to investigate whether adjuvant chemotherapy is essential for patients with early-stage serous and endometrioid epithelial ovarian cancer, the present study collected data from the US Surveillance, Epidemiology and End Results database between 2004 and 2015. All subjects underwent comprehensive staging surgery and were diagnosed as stages IA-IIA, grade 1–2. A total of 2644 patients were enrolled in the present study, among which 1589 patients received platinum-based chemotherapy. Comparisons of categorical data were performed via χ2 tests. Variables with P < 0.05 in univariate analyses were further analyzed using multiple logistic regression. Selection bias from the heterogeneity of demographic and clinical characteristics was avoided using propensity score matching. Cox proportional hazards models were applied to estimate hazard ratios (HRs) and 95% confidence intervals (CIs), investigating the association between variables and 5-year overall survival. After the propensity score matching, there was an equal number of patients with or without chemotherapy (n = 925). The results of the present study indicated that those aged ≥65 years were at an increased risk of ovarian cancer, and the age was associated with poor prognosis (HR, 1.486; CI, 1.208–1.827; P < 0.001). Endometrioid carcinoma was associated with improved 5-year overall survival compared with serous cystadenocarcinoma (HR, 0.697; CI, 0.584–0.833; P < 0.001). Chemotherapy could not prolong the 5-year overall survival of patients with early-stage serous and endometrioid ovarian cancer (HR, 1.092; CI, 0.954–1.249; P = 0.201). These results demonstrated that adjuvant chemotherapy was unnecessary for patients with early-stage serous and endometrioid ovarian cancer after they underwent comprehensive staging surgery.
Collapse
Affiliation(s)
- Shuqing Li
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 128 Shenyang Road, Shanghai, 200090, China
| | - Zhiling Zhu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 128 Shenyang Road, Shanghai, 200090, China.
| |
Collapse
|
10
|
Mlynska A, Vaišnorė R, Rafanavičius V, Jocys S, Janeiko J, Petrauskytė M, Bijeikis S, Cimmperman P, Intaitė B, Žilionytė K, Barakauskienė A, Meškauskas R, Paberalė E, Pašukonienė V. A gene signature for immune subtyping of desert, excluded, and inflamed ovarian tumors. Am J Reprod Immunol 2020; 84:e13244. [PMID: 32294293 DOI: 10.1111/aji.13244] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 03/24/2020] [Accepted: 04/07/2020] [Indexed: 12/19/2022] Open
Abstract
PROBLEM The current tumor immunology paradigm emphasizes the role of the immune tumor microenvironment and distinguishes several histologically and transcriptionally different immune tumor subtypes. However, the experimental validation of such classification is so far limited to selected cancer types. Here, we aimed to explore the existence of inflamed, excluded, and desert immune subtypes in ovarian cancer, as well as investigate their association with the disease outcome. METHOD OF STUDY We used the publicly available ovarian cancer dataset from The Cancer Genome Atlas for developing subtype assignment algorithm, which was next verified in a cohort of 32 real-world patients of a known tumor subtype. RESULTS Using clinical and gene expression data of 489 ovarian cancer patients in the publicly available dataset, we identified three transcriptionally distinct clusters, representing inflamed, excluded, and desert subtypes. We developed a two-step subtyping algorithm with COL5A2 serving as a marker for separating excluded tumors, and CD2, TAP1, and ICOS for distinguishing between inflamed and desert tumors. The accuracy of gene expression-based subtyping algorithm in a real-world cohort was 75%. Additionally, we confirmed that patients bearing inflamed tumors are more likely to survive longer. CONCLUSION Our results highlight the presence of transcriptionally and histologically distinct immune subtypes among ovarian tumors and emphasize the potential benefit of immune subtyping as a clinical tool for treatment tailoring.
Collapse
Affiliation(s)
| | | | | | - Simonas Jocys
- Baltic Institute of Advanced Technology, Vilnius, Lithuania
| | - Julija Janeiko
- Baltic Institute of Advanced Technology, Vilnius, Lithuania
| | | | - Simas Bijeikis
- Baltic Institute of Advanced Technology, Vilnius, Lithuania
| | | | | | | | - Aušrinė Barakauskienė
- Vilnius University, Vilnius, Lithuania.,Ltd Patologijos Diagnostika, Vilnius, Lithuania
| | | | | | | |
Collapse
|
11
|
Swayden M, Soubeyran P, Iovanna J. Upcoming Revolutionary Paths in Preclinical Modeling of Pancreatic Adenocarcinoma. Front Oncol 2020; 9:1443. [PMID: 32038993 PMCID: PMC6987422 DOI: 10.3389/fonc.2019.01443] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
To date, PDAC remains the cancer having the worst prognosis with mortality rates constantly on the rise. Efficient cures are still absent, despite all attempts to understand the aggressive physiopathology underlying this disease. A major stumbling block is the outdated preclinical modeling strategies applied in assessing effectiveness of novel anticancer therapeutics. Current in vitro preclinical models have a low fidelity to mimic the exact architectural and functional complexity of PDAC tumor found in human set, due to the lack of major components such as immune system and tumor microenvironment with its associated chemical and mechanical signals. The existing PDAC preclinical platforms are still far from being reliable and trustworthy to guarantee the success of a drug in clinical trials. Therefore, there is an urgent demand to innovate novel in vitro preclinical models that mirrors with precision tumor-microenvironment interface, pressure of immune system, and molecular and morphological aspects of the PDAC normally experienced within the living organ. This review outlines the traditional preclinical models of PDAC namely 2D cell lines, genetically engineered mice, and xenografts, and describing the present famous approach of 3D organoids. We offer a detailed narration of the pros and cons of each model system. Finally, we suggest the incorporation of two off-center newly born techniques named 3D bio-printing and organs-on-chip and discuss the potentials of swine models and in silico tools, as powerful new tools able to transform PDAC preclinical modeling to a whole new level and open new gates in personalized medicine.
Collapse
Affiliation(s)
- Mirna Swayden
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Philippe Soubeyran
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| |
Collapse
|
12
|
Kaipio K, Chen P, Roering P, Huhtinen K, Mikkonen P, Östling P, Lehtinen L, Mansuri N, Korpela T, Potdar S, Hynninen J, Auranen A, Grénman S, Wennerberg K, Hautaniemi S, Carpén O. ALDH1A1-related stemness in high-grade serous ovarian cancer is a negative prognostic indicator but potentially targetable by EGFR/mTOR-PI3K/aurora kinase inhibitors. J Pathol 2019; 250:159-169. [PMID: 31595974 DOI: 10.1002/path.5356] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 09/05/2019] [Accepted: 10/03/2019] [Indexed: 12/16/2022]
Abstract
Poor chemotherapy response remains a major treatment challenge for high-grade serous ovarian cancer (HGSC). Cancer stem cells are the major contributors to relapse and treatment failure as they can survive conventional therapy. Our objectives were to characterise stemness features in primary patient-derived cell lines, correlate stemness markers with clinical outcome and test the response of our cells to both conventional and exploratory drugs. Tissue and ascites samples, treatment-naive and/or after neoadjuvant chemotherapy, were prospectively collected. Primary cancer cells, cultured under conditions favouring either adherent or spheroid growth, were tested for stemness markers; the same markers were analysed in tissue and correlated with chemotherapy response and survival. Drug sensitivity and resistance testing was performed with 306 oncology compounds. Spheroid growth condition HGSC cells showed increased stemness marker expression (including aldehyde dehydrogenase isoform I; ALDH1A1) as compared with adherent growth condition cells, and increased resistance to platinum and taxane. A set of eight stemness markers separated treatment-naive tumours into two clusters and identified a distinct subgroup of HGSC with enriched stemness features. Expression of ALDH1A1, but not most other stemness markers, was increased after neoadjuvant chemotherapy and its expression in treatment-naive tumours correlated with chemoresistance and reduced survival. In drug sensitivity and resistance testing, five compounds, including two PI3K-mTOR inhibitors, demonstrated significant activity in both cell culture conditions. Thirteen compounds, including EGFR, PI3K-mTOR and aurora kinase inhibitors, were more toxic to spheroid cells than adherent cells. Our results identify stemness markers in HGSC that are associated with a decreased response to conventional chemotherapy and reduced survival if expressed by treatment-naive tumours. EGFR, mTOR-PI3K and aurora kinase inhibitors are candidates for targeting this cell population. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Katja Kaipio
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ping Chen
- Integrated Cardio Metabolic Centre (ICMC), Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Pia Roering
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kaisa Huhtinen
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Piia Mikkonen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Päivi Östling
- Science for Life Laboratory Department of Oncology & Pathology, Karolinska Institutet, Huddinge, Sweden.,Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Laura Lehtinen
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Naziha Mansuri
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Taina Korpela
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland, High Throughput Biomedicine Unit (HTB), University of Helsinki, Helsinki, Finland
| | - Johanna Hynninen
- Department of Obstetrics and Gynaecology, University of Turku and Turku University Hospital, Turku, Finland
| | - Annika Auranen
- Department of Obstetrics and Gynaecology, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Seija Grénman
- Department of Obstetrics and Gynaecology, University of Turku and Turku University Hospital, Turku, Finland
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland, High Throughput Biomedicine Unit (HTB), University of Helsinki, Helsinki, Finland.,Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Sampsa Hautaniemi
- Research Programs Unit, Genome-Scale Biology and Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olli Carpén
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland.,Research Programs Unit, Genome-Scale Biology and Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
13
|
Machine learning and data mining frameworks for predicting drug response in cancer: An overview and a novel in silico screening process based on association rule mining. Pharmacol Ther 2019; 203:107395. [DOI: 10.1016/j.pharmthera.2019.107395] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022]
|
14
|
Niu Y, Sun W, Chen K, Fu Z, Chen Y, Zhu J, Chen H, Shi Y, Zhang H, Wang L, Shen HM, Xia D, Wu Y. A Novel Scoring System for Pivotal Autophagy-Related Genes Predicts Outcomes after Chemotherapy in Advanced Ovarian Cancer Patients. Cancer Epidemiol Biomarkers Prev 2019; 28:2106-2114. [PMID: 31533939 DOI: 10.1158/1055-9965.epi-19-0359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/30/2019] [Accepted: 09/12/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In the clinical practice of ovarian cancer, the application of autophagy, an important regulator of carcinogenesis and chemoresistance, is still limited. This study aimed to establish a scoring system based on expression profiles of pivotal autophagy-related (ATG) genes in patients with stage III/IV ovarian cancer who received chemotherapy. METHODS Data of ovarian serous cystadenocarcinoma in The Cancer Genome Atlas (TCGA-OV) were used as training dataset. Two validation datasets comprised patients in a Chinese local database and a dataset from the Gene Expression Omnibus (GEO). ATG genes significantly (P < 0.1) associated with overall survival (OS) were selected and aggregated into an ATG scoring scale, of which the abilities to predict OS and recurrence-free survival (RFS) were examined. RESULTS Forty-three ATG genes were selected to develop the ATG score. In TCGA-OV, patients with lower ATG scores had better OS [HR = 0.41; 95% confidence interval (CI), 0.26-0.65; P < 0.001] and RFS [HR = 0.47; 95% CI, 0.27-0.82; P = 0.007]. After complete or partial remission to primary therapy, the rate of recurrence was 47.2% in the low-score group and 68.3% in the high-score group (odds ratio = 0.42; 95% CI, 0.18-0.92; P = 0.03). Such findings were verified in the two validation datasets. CONCLUSIONS We established a novel scoring system based on pivotal ATG genes, which accurately predicts the outcomes of patients with advanced ovarian cancer after chemotherapy. IMPACT The present ATG scoring system may provide a novel perspective and a promising tool for the development of personalized therapy in the future.
Collapse
Affiliation(s)
- Yuequn Niu
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjie Sun
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelie Chen
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiqin Fu
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yaqing Chen
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jianqing Zhu
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Hanwen Chen
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Honghe Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| | - Liming Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Dajing Xia
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yihua Wu
- Department of Toxicology of School of Public Health, and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
15
|
Chon HS, Sehovic M, Marchion D, Walko C, Xiong Y, Extermann M. Biologic Mechanisms Linked to Prognosis in Ovarian Cancer that May Be Affected by Aging. J Cancer 2019; 10:2604-2618. [PMID: 31258768 PMCID: PMC6584919 DOI: 10.7150/jca.29611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 04/27/2019] [Indexed: 12/20/2022] Open
Abstract
The increase of both life expectancy of the Western industrialized population and cancer incidence with aging is expected to result in a rapid expansion of the elderly cancer population, including patients with epithelial ovarian cancer (EOC). Although the survival of patients with EOC has generally improved over the past three decades, this progress has yet to provide benefits for elderly patients. Compared with young age, advanced age has been reported as an adverse prognostic factor influencing EOC. However, contradicting results have been obtained, and the mechanisms underlying this observation are poorly defined. Few papers have been published on the underlying biological mechanisms that might explain this prognosis trend. We provide an extensive review of mechanisms that have been linked to EOC prognosis and/or aging in the published literature and might underlie this relationship in humans.
Collapse
Affiliation(s)
- Hye Sook Chon
- Department of Gynecology Oncology, Moffitt Cancer Center and Research Institute, Tampa FL, USA
- University of South Florida, Tampa FL, USA
| | - Marina Sehovic
- Senior Adult Oncology Program, Moffitt Cancer Center and Research Institute, Tampa FL, USA
- Department of Individualized Cancer Management, Moffitt Cancer Center and Research Institute, Tampa FL, USA
| | - Douglas Marchion
- Department of Pathology, Moffitt Cancer Center and Research Institute, Tampa FL, USA
| | - Christine Walko
- Department of Individualized Cancer Management, Moffitt Cancer Center and Research Institute, Tampa FL, USA
| | - Yin Xiong
- Department of Pathology, Moffitt Cancer Center and Research Institute, Tampa FL, USA
| | - Martine Extermann
- Senior Adult Oncology Program, Moffitt Cancer Center and Research Institute, Tampa FL, USA
- Department of Individualized Cancer Management, Moffitt Cancer Center and Research Institute, Tampa FL, USA
- University of South Florida, Tampa FL, USA
| |
Collapse
|
16
|
Increased STAT1 Expression in High Grade Serous Ovarian Cancer Is Associated With a Better Outcome. Int J Gynecol Cancer 2019; 28:459-465. [PMID: 29303938 DOI: 10.1097/igc.0000000000001193] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Recently it has been demonstrated that constitutively activated signal transducer and activator of transcription 1 (STAT1) gene expression may act as a biomarker of ovarian cancer chemotherapy response. In this study, our objective was to validate the use of STAT1 immunohistochemistry as a prognostic biomarker for disease outcome using a cohort derived from Latin America. METHODS We evaluated a cohort of Brazilian high-grade serous ovarian cancer, comprising 65 patients with outcome data covering more than 5 years to determine the prognostic and predictive value of STAT1 expression levels. High-grade serous ovarian cancer tumors were used to construct a tissue microarray. Exploratory analyses were conducted on clinical, histopathological, and STAT1 expression data that included descriptive statistics and Pearson correlative analyses. Survival curves for disease-free survival and overall survival were obtained by the Kaplan-Meier method, and the significance of homogeneity between the classes was assessed by log-rank statistics (Mantel-Cox). RESULTS High expression of STAT1 in tumors was significantly associated with improved disease-free survival (P = 0.0256) and overall survival (P = 0.0193). Proportional hazards regression analysis showed STAT1 expression had an independent effect on both disease-free survival (P = 0.0358) and overall survival (P = 0.0469). CONCLUSIONS These findings from a Brazilian cohort of patients with ovarian cancer reinforce the association of high STAT1 expression with better response to chemotherapy, providing additional validation of this protein as both a prognostic and predictive biomarker. Collectively, these results together with other recently published studies increase the feasibility of using the STAT1 pathway for the development of novel immunomodulator drugs that could enhance response to treatment.
Collapse
|
17
|
Lu H, Arshad M, Thornton A, Avesani G, Cunnea P, Curry E, Kanavati F, Liang J, Nixon K, Williams ST, Hassan MA, Bowtell DDL, Gabra H, Fotopoulou C, Rockall A, Aboagye EO. A mathematical-descriptor of tumor-mesoscopic-structure from computed-tomography images annotates prognostic- and molecular-phenotypes of epithelial ovarian cancer. Nat Commun 2019; 10:764. [PMID: 30770825 PMCID: PMC6377605 DOI: 10.1038/s41467-019-08718-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 01/24/2019] [Indexed: 12/11/2022] Open
Abstract
The five-year survival rate of epithelial ovarian cancer (EOC) is approximately 35-40% despite maximal treatment efforts, highlighting a need for stratification biomarkers for personalized treatment. Here we extract 657 quantitative mathematical descriptors from the preoperative CT images of 364 EOC patients at their initial presentation. Using machine learning, we derive a non-invasive summary-statistic of the primary ovarian tumor based on 4 descriptors, which we name "Radiomic Prognostic Vector" (RPV). RPV reliably identifies the 5% of patients with median overall survival less than 2 years, significantly improves established prognostic methods, and is validated in two independent, multi-center cohorts. Furthermore, genetic, transcriptomic and proteomic analysis from two independent datasets elucidate that stromal phenotype and DNA damage response pathways are activated in RPV-stratified tumors. RPV and its associated analysis platform could be exploited to guide personalized therapy of EOC and is potentially transferrable to other cancer types.
Collapse
Affiliation(s)
- Haonan Lu
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Mubarik Arshad
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Andrew Thornton
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Giacomo Avesani
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Paula Cunnea
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Ed Curry
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Fahdi Kanavati
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Jack Liang
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Katherine Nixon
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Sophie T Williams
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Mona Ali Hassan
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - David D L Bowtell
- Peter MacCallum Cancer Centre, Melbourne, 3010, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, 3010, VIC, Australia
| | - Hani Gabra
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
- Early Clinical Development, iMED Biotech Unit, AstraZeneca, Cambridge, SG8 6HB, UK
| | - Christina Fotopoulou
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Andrea Rockall
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
- Department of Radiology, Imperial College Healthcare NHS Trust, London, W12 0HS, UK
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Eric O Aboagye
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK.
| |
Collapse
|
18
|
Losi L, Fonda S, Saponaro S, Chelbi ST, Lancellotti C, Gozzi G, Alberti L, Fabbiani L, Botticelli L, Benhattar J. Distinct DNA Methylation Profiles in Ovarian Tumors: Opportunities for Novel Biomarkers. Int J Mol Sci 2018; 19:ijms19061559. [PMID: 29882921 PMCID: PMC6032431 DOI: 10.3390/ijms19061559] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 01/16/2023] Open
Abstract
Aberrant methylation of multiple promoter CpG islands could be related to the biology of ovarian tumors and its determination could help to improve treatment strategies. DNA methylation profiling was performed using the Methylation Ligation-dependent Macroarray (MLM), an array-based analysis. Promoter regions of 41 genes were analyzed in 102 ovarian tumors and 17 normal ovarian samples. An average of 29% of hypermethylated promoter genes was observed in normal ovarian tissues. This percentage increased slightly in serous, endometrioid, and mucinous carcinomas (32%, 34%, and 45%, respectively), but decreased in germ cell tumors (20%). Ovarian tumors had methylation profiles that were more heterogeneous than other epithelial cancers. Unsupervised hierarchical clustering identified four groups that are very close to the histological subtypes of ovarian tumors. Aberrant methylation of three genes (BRCA1, MGMT, and MLH1), playing important roles in the different DNA repair mechanisms, were dependent on the tumor subtype and represent powerful biomarkers for precision therapy. Furthermore, a promising relationship between hypermethylation of MGMT, OSMR, ESR1, and FOXL2 and overall survival was observed. Our study of DNA methylation profiling indicates that the different histotypes of ovarian cancer should be treated as separate diseases both clinically and in research for the development of targeted therapies.
Collapse
Affiliation(s)
- Lorena Losi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy.
- Unit of Pathology, Azienda Ospedaliero-Universitaria Policlinico, 41124 Modena, Italy.
| | - Sergio Fonda
- Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy.
| | - Sara Saponaro
- Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy.
- Institute of Pathology, Lausanne University Hospital, 1011 Lausanne, Switzerland.
| | - Sonia T Chelbi
- Institute of Pathology, Lausanne University Hospital, 1011 Lausanne, Switzerland.
| | - Cesare Lancellotti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy.
| | - Gaia Gozzi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy.
| | - Loredana Alberti
- Institute of Pathology, Lausanne University Hospital, 1011 Lausanne, Switzerland.
| | - Luca Fabbiani
- Unit of Pathology, Azienda Ospedaliero-Universitaria Policlinico, 41124 Modena, Italy.
| | - Laura Botticelli
- Unit of Pathology, Azienda Ospedaliero-Universitaria Policlinico, 41124 Modena, Italy.
| | - Jean Benhattar
- Institute of Pathology, Lausanne University Hospital, 1011 Lausanne, Switzerland.
- Aurigen, Centre de Génétique et Pathologie, 1004 Lausanne, Switzerland.
| |
Collapse
|
19
|
Xu S, Yang Z, Jin P, Yang X, Li X, Wei X, Wang Y, Long S, Zhang T, Chen G, Sun C, Ma D, Gao Q. Metformin Suppresses Tumor Progression by Inactivating Stromal Fibroblasts in Ovarian Cancer. Mol Cancer Ther 2018; 17:1291-1302. [PMID: 29545331 DOI: 10.1158/1535-7163.mct-17-0927] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/21/2017] [Accepted: 03/06/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Sen Xu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zongyuan Yang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Jin
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Yang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoting Li
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiao Wei
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ya Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sixiang Long
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Taoran Zhang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Gang Chen
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chaoyang Sun
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ding Ma
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qinglei Gao
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
20
|
Hyter S, Hirst J, Pathak H, Pessetto ZY, Koestler DC, Raghavan R, Pei D, Godwin AK. Developing a genetic signature to predict drug response in ovarian cancer. Oncotarget 2018; 9:14828-14848. [PMID: 29599910 PMCID: PMC5871081 DOI: 10.18632/oncotarget.23663] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/13/2017] [Indexed: 12/15/2022] Open
Abstract
There is a lack of personalized treatment options for women with recurrent platinum-resistant ovarian cancer. Outside of bevacizumab and a group of poly ADP-ribose polymerase inhibitors, few options are available to women that relapse. We propose that efficacious drug combinations can be determined via molecular characterization of ovarian tumors along with pre-established pharmacogenomic profiles of repurposed compounds. To that end, we selectively performed multiple two-drug combination treatments in ovarian cancer cell lines that included reactive oxygen species inducers and HSP90 inhibitors. This allowed us to select cell lines that exhibit disparate phenotypes of proliferative inhibition to a specific drug combination of auranofin and AUY922. We profiled altered mechanistic responses from these agents in both reactive oxygen species and HSP90 pathways, as well as investigated PRKCI and lncRNA expression in ovarian cancer cell line models. Generation of dual multi-gene panels implicated in resistance or sensitivity to this drug combination was produced using RNA sequencing data and the validity of the resistant signature was examined using high-density RT-qPCR. Finally, data mining for the prevalence of these signatures in a large-scale clinical study alluded to the prevalence of resistant genes in ovarian tumor biology. Our results demonstrate that high-throughput viability screens paired with reliable in silico data can promote the discovery of effective, personalized therapeutic options for a currently untreatable disease.
Collapse
Affiliation(s)
- Stephen Hyter
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeff Hirst
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Harsh Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ziyan Y. Pessetto
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Devin C. Koestler
- University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Rama Raghavan
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Dong Pei
- University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew K. Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
21
|
Xu K, Yang S, Zhao Y. Prognostic significance of BRCA mutations in ovarian cancer: an updated systematic review with meta-analysis. Oncotarget 2018; 8:285-302. [PMID: 27690218 PMCID: PMC5352118 DOI: 10.18632/oncotarget.12306] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/22/2016] [Indexed: 12/12/2022] Open
Abstract
There is no consensus on the syntheses concerning the impact of BRCA mutation on ovarian cancer survival. A systematic review and meta-analysis of observational studies was conducted that evaluated the impact of BRCA mutations on the survival outcomes of patients with ovarian cancer. The primary outcome measure was overall survival (OS) and secondary outcome was progression-free survival (PFS). We presented data with hazard ratios (HRs) and 95% confidence interval (CI) and pooled them using the random-effects models. From 2,624 unique records, 34 eligible studies including 18,396 patients were identified. BRCA1/2 mutations demonstrated both OS and PFS benefits in patients with ovarian cancer (OS: HR = 0.67, 95% CI, 0.57 to 0.78, I2 = 76.5%, P <0.001; PFS: HR = 0.62, 95% CI, 0.53 to 0.73, I2 = 18.1%, P = 0.261). For BRCA1 mutation carriers, the HRs for OS and PFS benefits were 0.73 (95% CI, 0.63 to 0.86) and 0.68 (95% CI, 0.52 to 0.89), respectively. For BRCA2 mutation carriers, the HRs for OS and PFS benefits were 0.57 (95% CI, 0.45 to 0.73) and 0.48 (95% CI, 0.30 to 0.75), respectively. The results of subgroup analyses for OS stratified by study quality, tumor stage, study design, sample size, number of research center, duration of follow-up, baseline characteristics adjusted and tumor histology were mostly constant across BRCA1/2, BRCA1 and BRCA2 mutation subtypes. In summary, for patients with ovarian cancer, BRCA mutations were associated with improved OS and PFS. Further large-scale prospective cohort studies should be conducted to test its benefits in specific patients.
Collapse
Affiliation(s)
- Kai Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shouhua Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yingchao Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
22
|
Azuaje F. Computational models for predicting drug responses in cancer research. Brief Bioinform 2017; 18:820-829. [PMID: 27444372 PMCID: PMC5862310 DOI: 10.1093/bib/bbw065] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Indexed: 02/06/2023] Open
Abstract
The computational prediction of drug responses based on the analysis of multiple types of genome-wide molecular data is vital for accomplishing the promise of precision medicine in oncology. This will benefit cancer patients by matching their tumor characteristics to the most effective therapy available. As larger and more diverse layers of patient-related data become available, further demands for new bioinformatics approaches and expertise will arise. This article reviews key strategies, resources and techniques for the prediction of drug sensitivity in cell lines and patient-derived samples. It discusses major advances and challenges associated with the different model development steps. This review highlights major trends in this area, and will assist researchers in the assessment of recent progress and in the selection of approaches to emerging applications in oncology.
Collapse
Affiliation(s)
- Francisco Azuaje
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg
- Corresponding author: Francisco Azuaje, NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health (LIH), Luxembourg L-1526, Luxembourg. Tel.: +352-26970875; Fax: +352-26970396; E-mail:
| |
Collapse
|
23
|
Lund RJ, Huhtinen K, Salmi J, Rantala J, Nguyen EV, Moulder R, Goodlett DR, Lahesmaa R, Carpén O. DNA methylation and Transcriptome Changes Associated with Cisplatin Resistance in Ovarian Cancer. Sci Rep 2017; 7:1469. [PMID: 28473707 PMCID: PMC5431431 DOI: 10.1038/s41598-017-01624-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/30/2017] [Indexed: 12/31/2022] Open
Abstract
High-grade serous ovarian cancer is the most common ovarian cancer type. Although the combination of surgery and platinum-taxane chemotherapy provide an effective treatment, drug resistance frequently occurs leading to poor outcome. In order to clarify the molecular mechanisms of drug resistance, the DNA methylation and transcriptomic changes, associated with the development of drug resistance in high-grade serous ovarian cancer, were examined from patient derived malignant ascites cells. In parallel with large-scale transcriptome changes, cisplatin resistance was associated with loss of hypermethylation at several CpG sites primarily localized in the intergenic regions of the genome. The transcriptome and CpG methylome changes in response to cisplatin treatment of both sensitive and resistant cells were minimal, indicating the importance of post-translational mechanisms in regulating death or survival of the cells. The response of resistant cells to high concentrations of cisplatin revealed transcriptomic changes in potential key drivers of drug resistance, such as KLF4. Among the strongest changes was also induction of IL6 in resistant cells and the expression was further increased in response to cisplatin. Also, several other components of IL6 signaling were affected, further supporting previous observations on its importance in malignant transformation and development of drug resistance in ovarian cancer.
Collapse
Affiliation(s)
- Riikka J Lund
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.
| | - Kaisa Huhtinen
- Department of Pathology, Medicity Research Unit, University of Turku and Turku University Hospital, Turku, Finland
| | - Jussi Salmi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Juha Rantala
- Department of Pathology, Medicity Research Unit, University of Turku and Turku University Hospital, Turku, Finland
| | - Elizabeth V Nguyen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Robert Moulder
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - David R Goodlett
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, USA
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Olli Carpén
- Department of Pathology, Medicity Research Unit, University of Turku and Turku University Hospital, Turku, Finland.
| |
Collapse
|
24
|
Nguyen EV, Huhtinen K, Goo YA, Kaipio K, Andersson N, Rantanen V, Hynninen J, Lahesmaa R, Carpen O, Goodlett DR. Hyper-phosphorylation of Sequestosome-1 Distinguishes Resistance to Cisplatin in Patient Derived High Grade Serous Ovarian Cancer Cells. Mol Cell Proteomics 2017; 16:1377-1392. [PMID: 28455291 DOI: 10.1074/mcp.m116.058321] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/22/2017] [Indexed: 12/13/2022] Open
Abstract
Platinum-resistance is a major limitation to effective chemotherapy regimens in high-grade serous ovarian cancer (HGSOC). To better understand the mechanisms involved we characterized the proteome and phosphoproteome in cisplatin sensitive and resistant HGSOC primary cells using a mass spectrometry-based proteomic strategy. PCA analysis identified a distinctive phosphoproteomic signature between cisplatin sensitive and resistant cell lines. The most phosphorylated protein in cisplatin resistant cells was sequestosome-1 (p62/SQSTM1). Changes in expression of apoptosis and autophagy related proteins Caspase-3 and SQSTM1, respectively, were validated by Western blot analysis. A significant increase in apoptosis in the presence of cisplatin was observed in only the sensitive cell line while SQSTM1 revealed increased expression in the resistant cell line relative to sensitive cell line. Furthermore, site-specific phosphorylation on 20 amino acid residues of SQSTM1 was detected indicating a hyper-phosphorylation phenotype. This elevated hyper-phosphorylation of SQSTM1 in resistant HGSOC cell lines was validated with Western blot analysis. Immunofluoresence staining of s28-pSQSTM1 showed inducible localization to autophagosomes upon cisplatin treatment in the sensitive cell line while being constitutively expressed to autophagosomes in the resistant cell. Furthermore, SQSTM1 expression was localized in cancer cells of clinical high-grade serous tumors. Here, we propose hyper-phosphorylation of SQSTM1 as a marker and a key proteomic change in cisplatin resistance development in ovarian cancers by activating the autophagy pathway and influencing down-regulation of apoptosis.
Collapse
Affiliation(s)
- Elizabeth V Nguyen
- From the ‡Turku Centre of Biotechnology, University of Turku and Åbo Akademi, Tykistökatu 6, Turku 20520, Finland.,§Department of Pathology, Medicity Research Unit, University of Turku and Turku University Hospital, Tykistökatu 6, Turku 20520, Finland
| | - Kaisa Huhtinen
- §Department of Pathology, Medicity Research Unit, University of Turku and Turku University Hospital, Tykistökatu 6, Turku 20520, Finland.,§Department of Pathology, Medicity Research Unit, University of Turku and Turku University Hospital, Tykistökatu 6, Turku 20520, Finland
| | - Young Ah Goo
- ¶Department of Pharmaceutical Sciences, University of Maryland, 20 North Pine Street, Room N707, Maryland 21201
| | - Katja Kaipio
- §Department of Pathology, Medicity Research Unit, University of Turku and Turku University Hospital, Tykistökatu 6, Turku 20520, Finland
| | - Noora Andersson
- ‖Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Haartmaninkatu 3, 00290 Helsinki, Finland
| | - Ville Rantanen
- **Research Programs Unit, Genome-Scale Biology, Medicum and Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, POB 63, Helsinki, 00014 Finland
| | - Johanna Hynninen
- ‡‡Department of Obstetrics and Gynecology, University of Turku and Turku University Hospital, Kiinamyllynkatu 4-8, Turku 20521, Finland
| | - Riitta Lahesmaa
- From the ‡Turku Centre of Biotechnology, University of Turku and Åbo Akademi, Tykistökatu 6, Turku 20520, Finland
| | - Olli Carpen
- §Department of Pathology, Medicity Research Unit, University of Turku and Turku University Hospital, Tykistökatu 6, Turku 20520, Finland.,‖Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Haartmaninkatu 3, 00290 Helsinki, Finland
| | - David R Goodlett
- From the ‡Turku Centre of Biotechnology, University of Turku and Åbo Akademi, Tykistökatu 6, Turku 20520, Finland; .,¶Department of Pharmaceutical Sciences, University of Maryland, 20 North Pine Street, Room N707, Maryland 21201
| |
Collapse
|
25
|
Xu K, Yang S, Zhao Y. Prognostic significance of BRCA mutations in ovarian cancer: an updated systematic review with meta-analysis. Oncotarget 2017. [PMID: 27690218 DOI: 10.18632/oncotarget.12306] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
There is no consensus on the syntheses concerning the impact of BRCA mutation on ovarian cancer survival. A systematic review and meta-analysis of observational studies was conducted that evaluated the impact of BRCA mutations on the survival outcomes of patients with ovarian cancer. The primary outcome measure was overall survival (OS) and secondary outcome was progression-free survival (PFS). We presented data with hazard ratios (HRs) and 95% confidence interval (CI) and pooled them using the random-effects models. From 2,624 unique records, 34 eligible studies including 18,396 patients were identified. BRCA1/2 mutations demonstrated both OS and PFS benefits in patients with ovarian cancer (OS: HR = 0.67, 95% CI, 0.57 to 0.78, I2 = 76.5%, P <0.001; PFS: HR = 0.62, 95% CI, 0.53 to 0.73, I2 = 18.1%, P = 0.261). For BRCA1 mutation carriers, the HRs for OS and PFS benefits were 0.73 (95% CI, 0.63 to 0.86) and 0.68 (95% CI, 0.52 to 0.89), respectively. For BRCA2 mutation carriers, the HRs for OS and PFS benefits were 0.57 (95% CI, 0.45 to 0.73) and 0.48 (95% CI, 0.30 to 0.75), respectively. The results of subgroup analyses for OS stratified by study quality, tumor stage, study design, sample size, number of research center, duration of follow-up, baseline characteristics adjusted and tumor histology were mostly constant across BRCA1/2, BRCA1 and BRCA2 mutation subtypes. In summary, for patients with ovarian cancer, BRCA mutations were associated with improved OS and PFS. Further large-scale prospective cohort studies should be conducted to test its benefits in specific patients.
Collapse
Affiliation(s)
- Kai Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shouhua Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yingchao Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
26
|
Xu K, Yang S, Zhao Y. Prognostic significance of BRCA mutations in ovarian cancer: an updated systematic review with meta-analysis. Oncotarget 2017. [PMID: 27690218 DOI: 10.18632/oncotarget.12306]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
There is no consensus on the syntheses concerning the impact of BRCA mutation on ovarian cancer survival. A systematic review and meta-analysis of observational studies was conducted that evaluated the impact of BRCA mutations on the survival outcomes of patients with ovarian cancer. The primary outcome measure was overall survival (OS) and secondary outcome was progression-free survival (PFS). We presented data with hazard ratios (HRs) and 95% confidence interval (CI) and pooled them using the random-effects models. From 2,624 unique records, 34 eligible studies including 18,396 patients were identified. BRCA1/2 mutations demonstrated both OS and PFS benefits in patients with ovarian cancer (OS: HR = 0.67, 95% CI, 0.57 to 0.78, I2 = 76.5%, P <0.001; PFS: HR = 0.62, 95% CI, 0.53 to 0.73, I2 = 18.1%, P = 0.261). For BRCA1 mutation carriers, the HRs for OS and PFS benefits were 0.73 (95% CI, 0.63 to 0.86) and 0.68 (95% CI, 0.52 to 0.89), respectively. For BRCA2 mutation carriers, the HRs for OS and PFS benefits were 0.57 (95% CI, 0.45 to 0.73) and 0.48 (95% CI, 0.30 to 0.75), respectively. The results of subgroup analyses for OS stratified by study quality, tumor stage, study design, sample size, number of research center, duration of follow-up, baseline characteristics adjusted and tumor histology were mostly constant across BRCA1/2, BRCA1 and BRCA2 mutation subtypes. In summary, for patients with ovarian cancer, BRCA mutations were associated with improved OS and PFS. Further large-scale prospective cohort studies should be conducted to test its benefits in specific patients.
Collapse
Affiliation(s)
- Kai Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shouhua Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yingchao Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
27
|
Treatment related toxicity in BRCA1-associated epithelial ovarian cancer - is DNA repairing impairment associated with more adverse events? Contemp Oncol (Pozn) 2016; 20:381-384. [PMID: 28373819 PMCID: PMC5371704 DOI: 10.5114/wo.2016.64597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 03/14/2016] [Indexed: 11/17/2022] Open
Abstract
Aim of the study The presence of BRCA germline mutations in patients with ovarian cancer has been shown to have predictive and prognostic significance, including increased platinum-sensitivity. The aim of the study was to evaluate if patients with BRCA1-associated ovarian cancer have more treatment related adverse events and, if so, does it have impact on chemotherapy outcomes. Material and methods We conducted a retrospective analysis of medical records of 172 patients with newly diagnosed epithelial ovarian cancer, treated in Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch between 2007 and 2013. Ninety-six of these patients have known BRCA mutation status – 21 patients were BRCA1(+) and 75 BRCA1(–). Analysed treatment related adverse events (AE’s) were: haematological toxicity, nausea/vomiting, neuropathy and mucositis. Results Grade 3–4 haematological AE’s were significantly more common among BRCA1(+) patients (OR = 3.86; 95% CI: 1.14–13.23; p = 0.02). There was no association between BRCA1 mutation status and neuropathy (p = 0.73) or nausea/vomiting (p = 0.91). Occurrence of above mentioned AE’s has no significant association with PFS (p = 0.75, 0.64, 0.97 respectively) and OS (p = 0.64, 0.69, 0.73 respectively). Conclusions Among patients with BRCA1-associated epithelial ovarian cancer we observed significantly more grade 3–4 haematological complications after chemotherapy. However, occurrence of AE’s did not correlate with better outcomes in this subgroup.
Collapse
|
28
|
Implementing rapid, robust, cost-effective, patient-centred, routine genetic testing in ovarian cancer patients. Sci Rep 2016; 6:29506. [PMID: 27406733 PMCID: PMC4942815 DOI: 10.1038/srep29506] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/17/2016] [Indexed: 01/09/2023] Open
Abstract
Advances in DNA sequencing have made genetic testing fast and affordable, but limitations of testing processes are impeding realisation of patient benefits. Ovarian cancer exemplifies the potential value of genetic testing and the shortcomings of current pathways to access testing. Approximately 15% of ovarian cancer patients have a germline BRCA1 or BRCA2 mutation which has substantial implications for their personal management and that of their relatives. Unfortunately, in most countries, routine implementation of BRCA testing for ovarian cancer patients has been inconsistent and largely unsuccessful. We developed a rapid, robust, mainstream genetic testing pathway in which testing is undertaken by the trained cancer team with cascade testing to relatives performed by the genetics team. 207 women with ovarian cancer were offered testing through the mainstream pathway. All accepted. 33 (16%) had a BRCA mutation. The result informed management of 79% (121/154) women with active disease. Patient and clinician feedback was very positive. The pathway offers a 4-fold reduction in time and 13-fold reduction in resource requirement compared to the conventional testing pathway. The mainstream genetic testing pathway we present is effective, efficient and patient-centred. It can deliver rapid, robust, large-scale, cost-effective genetic testing of BRCA1 and BRCA2 and may serve as an exemplar for other genes and other diseases.
Collapse
|
29
|
Au KK, Josahkian JA, Francis JA, Squire JA, Koti M. Current state of biomarkers in ovarian cancer prognosis. Future Oncol 2015; 11:3187-95. [DOI: 10.2217/fon.15.251] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
High-grade serous ovarian cancer remains one of the most lethal malignancies in women. Despite recent advances in surgical and pharmaceutical therapies, survival rates remain poor. A major impediment in management of this disease, that continues to contribute to poor overall survival rates, is resistance to standard carboplatin-paclitaxel combination chemotherapies. In addition to tumor cell intrinsic mechanisms leading to drug resistance, there is increasing awareness of the crucial role of the tumor microenvironment in mediating natural immune defense mechanisms and selective pressures that appear to facilitate chemotherapy sensitivity. We provide an overview of some of the promising new genetic and immunological biomarkers in ovarian cancer and discuss their biology and their likely clinical utility in future ovarian cancer management.
Collapse
Affiliation(s)
- Katrina K Au
- Department of Biomedical & Molecular Sciences, Queen's University, 99 University Ave., Kingston, ON, K7L 3N6, Canada
| | - Juliana A Josahkian
- Departments of Genetics & Pathology, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil
| | - Julie-Ann Francis
- Department of Obstetrics & Gynecology, Kingston General Hospital, 76 Stuart St, Kingston, ON, K7L 2V7, Canada
| | - Jeremy A Squire
- Departments of Genetics & Pathology, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil
| | - Madhuri Koti
- Department of Biomedical & Molecular Sciences, Queen's University, 99 University Ave., Kingston, ON, K7L 3N6, Canada
- Department of Obstetrics & Gynecology, Kingston General Hospital, 76 Stuart St, Kingston, ON, K7L 2V7, Canada
| |
Collapse
|
30
|
Alkema NG, Wisman GBA, van der Zee AGJ, van Vugt MATM, de Jong S. Studying platinum sensitivity and resistance in high-grade serous ovarian cancer: Different models for different questions. Drug Resist Updat 2015; 24:55-69. [PMID: 26830315 DOI: 10.1016/j.drup.2015.11.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/04/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022]
Abstract
High-grade serous ovarian cancer (HGSOC) has the highest mortality rate among all gynecological cancers. Patients are generally diagnosed in an advanced stage with the majority of cases displaying platinum resistant relapses. Recent genomic interrogation of large numbers of HGSOC patient samples indicated high complexity in terms of genetic aberrations, intra- and intertumor heterogeneity and underscored their lack of targetable oncogenic mutations. Sub-classifications of HGSOC based on expression profiles, termed 'differentiated', 'immunoreactive', 'mesenchymal' and 'proliferative', were shown to have prognostic value. In addition, in almost half of all HGSOC patients, a deficiency in homologous recombination (HR) was found that potentially can be targeted using PARP inhibitors. Developing precision medicine requires advanced experimental models. In the current review, we discuss experimental HGSOC models in which resistance to platinum therapy and the use of novel therapeutics can be carefully studied. Panels of better-defined primary cell lines need to be established to capture the full spectrum of HGSOC subtypes. Further refinement of cell lines is obtained with a 3-dimensional culture model mimicking the tumor microenvironment. Alternatively, ex vivo ovarian tumor tissue slices are used. For in vivo studies, larger panels of ovarian cancer patient-derived xenografts (PDXs) are being established, encompassing all expression subtypes. Ovarian cancer PDXs grossly retain tumor heterogeneity and clinical response to platinum therapy is preserved. PDXs are currently used in drug screens and as avatars for patient response. The role of the immune system in tumor responses can be assessed using humanized PDXs and immunocompetent genetically engineered mouse models. Dynamic tracking of genetic alterations in PDXs as well as patients during treatment and after relapse is feasible by sequencing circulating cell-free tumor DNA and analyzing circulating tumor cells. We discuss how various models and methods can be combined to delineate the molecular mechanisms underlying platinum resistance and to select HGSOC patients other than BRCA1/2-mutation carriers that could potentially benefit from the synthetic lethality of PARP inhibitors. This integrated approach is a first step to improve therapy outcomes in specific subgroups of HGSOC patients.
Collapse
Affiliation(s)
- Nicolette G Alkema
- Department of Gynecologic Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G Bea A Wisman
- Department of Gynecologic Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ate G J van der Zee
- Department of Gynecologic Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marcel A T M van Vugt
- Department of Medical Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Steven de Jong
- Department of Medical Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|