1
|
Dakal TC, Dhabhai B, Pant A, Moar K, Chaudhary K, Yadav V, Ranga V, Sharma NK, Kumar A, Maurya PK, Maciaczyk J, Schmidt‐Wolf IGH, Sharma A. Oncogenes and tumor suppressor genes: functions and roles in cancers. MedComm (Beijing) 2024; 5:e582. [PMID: 38827026 PMCID: PMC11141506 DOI: 10.1002/mco2.582] [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: 09/18/2023] [Revised: 04/21/2024] [Accepted: 04/26/2024] [Indexed: 06/04/2024] Open
Abstract
Cancer, being the most formidable ailment, has had a profound impact on the human health. The disease is primarily associated with genetic mutations that impact oncogenes and tumor suppressor genes (TSGs). Recently, growing evidence have shown that X-linked TSGs have specific role in cancer progression and metastasis as well. Interestingly, our genome harbors around substantial portion of genes that function as tumor suppressors, and the X chromosome alone harbors a considerable number of TSGs. The scenario becomes even more compelling as X-linked TSGs are adaptive to key epigenetic processes such as X chromosome inactivation. Therefore, delineating the new paradigm related to X-linked TSGs, for instance, their crosstalk with autosome and involvement in cancer initiation, progression, and metastasis becomes utmost importance. Considering this, herein, we present a comprehensive discussion of X-linked TSG dysregulation in various cancers as a consequence of genetic variations and epigenetic alterations. In addition, the dynamic role of X-linked TSGs in sex chromosome-autosome crosstalk in cancer genome remodeling is being explored thoroughly. Besides, the functional roles of ncRNAs, role of X-linked TSG in immunomodulation and in gender-based cancer disparities has also been highlighted. Overall, the focal idea of the present article is to recapitulate the findings on X-linked TSG regulation in the cancer landscape and to redefine their role toward improving cancer treatment strategies.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Department of BiotechnologyGenome and Computational Biology LabMohanlal Sukhadia UniversityUdaipurRajasthanIndia
| | - Bhanupriya Dhabhai
- Department of BiotechnologyGenome and Computational Biology LabMohanlal Sukhadia UniversityUdaipurRajasthanIndia
| | - Anuja Pant
- Department of BiochemistryCentral University of HaryanaMahendergarhHaryanaIndia
| | - Kareena Moar
- Department of BiochemistryCentral University of HaryanaMahendergarhHaryanaIndia
| | - Kanika Chaudhary
- School of Life Sciences. Jawaharlal Nehru UniversityNew DelhiIndia
| | - Vikas Yadav
- School of Life Sciences. Jawaharlal Nehru UniversityNew DelhiIndia
| | - Vipin Ranga
- Dearptment of Agricultural BiotechnologyDBT‐NECAB, Assam Agricultural UniversityJorhatAssamIndia
| | | | - Abhishek Kumar
- Manipal Academy of Higher EducationManipalKarnatakaIndia
- Institute of Bioinformatics, International Technology ParkBangaloreIndia
| | - Pawan Kumar Maurya
- Department of BiochemistryCentral University of HaryanaMahendergarhHaryanaIndia
| | - Jarek Maciaczyk
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
| | - Ingo G. H. Schmidt‐Wolf
- Department of Integrated OncologyCenter for Integrated Oncology (CIO)University Hospital BonnBonnGermany
| | - Amit Sharma
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
- Department of Integrated OncologyCenter for Integrated Oncology (CIO)University Hospital BonnBonnGermany
| |
Collapse
|
2
|
Zhang Y, Li D. An original aneuploidy-related gene model for predicting lung adenocarcinoma survival and guiding therapy. Sci Rep 2024; 14:8135. [PMID: 38584220 PMCID: PMC10999435 DOI: 10.1038/s41598-024-58020-y] [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: 11/02/2023] [Accepted: 03/25/2024] [Indexed: 04/09/2024] Open
Abstract
Aneuploidy is a hallmark of cancers, but the role of aneuploidy-related genes in lung adenocarcinoma (LUAD) and their prognostic value remain elusive. Gene expression and copy number variation (CNV) data were enrolled from TCGA and GEO database. Consistency clustering analysis was performed for molecular cluster. Tumor microenvironment was assessed by the xCell and ESTIMATE algorithm. Limma package was used for selecting differentially expressed genes (DEGs). LASSO and stepwise multivariate Cox regression analysis were used to establish an aneuploidy-related riskscore (ARS) signature. GDSC database was conducted to predict drug sensitivity. A nomogram was designed by rms R package. TCGA-LUAD patients were stratified into 3 clusters based on CNV data. The C1 cluster displayed the optimal survival advantage and highest inflammatory infiltration. Based on integrated intersecting DEGs, we constructed a 6-gene ARS model, which showed effective prediction for patient's survival. Drug sensitivity test predicted possible sensitive drugs in two risk groups. Additionally, the nomogram exhibited great predictive clinical treatment benefits. We established a 6-gene aneuploidy-related signature that could effectively predict the survival and therapy for LUAD patients. Additionally, the ARS model and nomogram could offer guidance for the preoperative estimation and postoperative therapy of LUAD.
Collapse
Affiliation(s)
- Yalei Zhang
- Department of Thoracic Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510032, China.
| | - Dongmei Li
- Department of Thoracic Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510032, China
| |
Collapse
|
3
|
Richau CS, Scherer NDM, Matta BP, de Armas EM, de Barros Moreira FC, Bergmann A, Pereira Chaves CB, Boroni M, dos Santos ACE, Moreira MAM. BRCA1, BRCA2, and TP53 germline and somatic variants and clinicopathological characteristics of Brazilian patients with epithelial ovarian cancer. Cancer Med 2024; 13:e6729. [PMID: 38308422 PMCID: PMC10905552 DOI: 10.1002/cam4.6729] [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: 06/21/2023] [Revised: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Approximately 3/4 of ovarian cancers are diagnosed in advanced stages, with the high-grade epithelial ovarian carcinoma (EOC) accounting for 90% of the cases. EOC present high genomic instability and somatic loss-of-function variants in genes associated with homologous recombination mutational repair pathway (HR), such as BRCA1 and BRCA2, and in TP53. The identification of germline variants in HR genes in EOC is relevant for treatment of platinum resistant tumors and relapsed tumors with therapies based in synthetic lethality such as PARP inhibitors. Patients with somatic variants in HR genes may also benefit from these therapies. In this work was analyzed the frequency of somatic variants in BRCA1, BRCA2, and TP53 in an EOC cohort of Brazilian patients, estimating the proportion of variants in tumoral tissue and their association with progression-free survival and overall survival. METHODS The study was conducted with paired blood/tumor samples from 56 patients. Germline and tumoral sequences of BRCA1, BRCA2, and TP53 were obtained by massive parallel sequencing. The HaplotypeCaller method was used for calling germline variants, and somatic variants were called with Mutect2. RESULTS A total of 26 germline variants were found, and seven patients presented germline pathogenic or likely pathogenic variants in BRCA1 or BRCA2. The analysis of tumoral tissue identified 52 somatic variants in 41 patients, being 43 somatic variants affecting or likely affecting protein functionality. Survival analyses showed that tumor staging was associated with overall survival (OS), while the presence of somatic mutation in TP53 was not associated with OS or progression-free survival. CONCLUSION Frequency of pathogenic or likely pathogenic germline variants in BRCA1 and BRCA2 (12.5%) was lower in comparison with other studies. TP53 was the most altered gene in tumors, with 62.5% presenting likely non-functional or non-functional somatic variants, while eight 14.2% presented likely non-functional or non-functional somatic variants in BRCA1 or BRCA2.
Collapse
Affiliation(s)
| | | | - Bruna Palma Matta
- Tumoral Genetics and Virology ProgramInstituto Nacional de CâncerRio de JaneiroBrazil
- Present address:
Hospital BP ‐ A Beneficência Portuguesa de São PauloSão PauloBrazil
| | | | | | - Anke Bergmann
- Clinical EpidemiologyInstituto Nacional de CâncerRio de JaneiroBrazil
| | | | - Mariana Boroni
- Bioinformatics and Computational Biology LaboratoryInstituto Nacional de CâncerRio de JaneiroBrazil
| | | | | |
Collapse
|
4
|
Svoboda M, Lohajova Behulova R, Slamka T, Sebest L, Repiska V. Comprehensive Genomic Profiling in Predictive Testing of Cancer. Physiol Res 2023; 72:S267-S275. [PMID: 37888970 PMCID: PMC10669951 DOI: 10.33549/physiolres.935154] [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: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 12/01/2023] Open
Abstract
Despite the rapid progress in the field of personalized medicine and the efforts to apply specific treatment strategies to patients based on the presence of pathogenic variants in one, two, or three genes, patient response to the treatment in terms of positive benefit and overall survival remains heterogeneous. However, advances in sequencing and bioinformatics technologies have facilitated the simultaneous examination of somatic variants in tens to thousands of genes in tumor tissue, enabling the determination of personalized management based on the patient's comprehensive genomic profile (CGP). CGP has the potential to enhance clinical decision-making and personalize innovative treatments for individual patients, by providing oncologists with a more comprehensive molecular characterization of tumors. This study aimed to highlight the utility of CGP in routine clinical practice. Here we present three patient cases with various advanced cancer indicated for CGP analysis using a combination of SOPHiA Solid Tumor Solution (STS, 42 genes) for DNA and SOPHiA RNAtarget Oncology Solution (ROS, 45 genes and 17 gene fusions with any random partners) for RNA. We were able to identify actionable genomic alterations in all three cases, thereby presenting valuable information for future management of these patients. This approach has the potential to transform clinical practice and greatly improve patient outcomes in the field of oncology.
Collapse
Affiliation(s)
- M Svoboda
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic.
| | | | | | | | | |
Collapse
|
5
|
Song Z, Zhang J, Sun Y, Jiang Z, Liu X. Establishment and validation of an immune infiltration predictive model for ovarian cancer. BMC Med Genomics 2023; 16:227. [PMID: 37759229 PMCID: PMC10538244 DOI: 10.1186/s12920-023-01657-x] [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: 11/08/2022] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The most prevalent mutation in ovarian cancer is the TP53 mutation, which impacts the development and prognosis of the disease. We looked at how the TP53 mutation associates the immunophenotype of ovarian cancer and the prognosis of the disease. METHODS We investigated the state of TP53 mutations and expression profiles in culturally diverse groups and datasets and developed an immune infiltration predictive model relying on immune-associated genes differently expressed between TP53 WT and TP53 MUT ovarian cancer cases. We aimed to construct an immune infiltration predictive model (IPM) to enhance the prognosis of ovarian cancer and investigate the impact of the IPM on the immunological microenvironment. RESULTS TP53 mutagenesis affected the expression of seventy-seven immune response-associated genes. An IPM was implemented and evaluated on ovarian cancer patients to distinguish individuals with low- and high-IPM subgroups of poor survival. For diagnostic and therapeutic use, a nomogram is thus created. According to pathway enrichment analysis, the pathways of the human immune response and immune function abnormalities were the most associated functions and pathways with the IPM genes. Furthermore, patients in the high-risk group showed low proportions of macrophages M1, activated NK cells, CD8+ T cells, and higher CTLA-4, PD-1, PD-L1, and TIM-3 than patients in the low-risk group. CONCLUSIONS The IPM model may identify high-risk patients and integrate other clinical parameters to predict their overall survival, suggesting it is a potential methodology for optimizing ovarian cancer prognosis.
Collapse
Affiliation(s)
- Zhenxia Song
- Department of Obstetrics, Qingdao women and childeren's hospital, #6 Tongfu Road, Shibei District, Qingdao, Shandong, 266000, P. R. China
| | - Jingwen Zhang
- Department of Obstetrics, Qingdao women and childeren's hospital, #6 Tongfu Road, Shibei District, Qingdao, Shandong, 266000, P. R. China
| | - Yue Sun
- Department of Obstetrics, Qingdao women and childeren's hospital, #6 Tongfu Road, Shibei District, Qingdao, Shandong, 266000, P. R. China
| | - Zhongmin Jiang
- Department of Pathology, Tian Jin Fifth's Central Hospital, #41 Zhejiang Road, Binhai District, Tianjin, 300450, P. R. China
| | - Xiaoning Liu
- Department of Obstetrics, Qingdao women and childeren's hospital, #6 Tongfu Road, Shibei District, Qingdao, Shandong, 266000, P. R. China.
| |
Collapse
|
6
|
Predicting Prognosis and Platinum Resistance in Ovarian Cancer: Role of Immunohistochemistry Biomarkers. Int J Mol Sci 2023; 24:ijms24031973. [PMID: 36768291 PMCID: PMC9916805 DOI: 10.3390/ijms24031973] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Ovarian cancer is a lethal reproductive tumour affecting women worldwide. The advancement in presentation and occurrence of chemoresistance are the key factors for poor survival among ovarian cancer women. Surgical debulking was the mainstay of systemic treatment for ovarian cancer, which was followed by a successful start to platinum-based chemotherapy. However, most women develop platinum resistance and relapse within six months of receiving first-line treatment. Thus, there is a great need to identify biomarkers to predict platinum resistance before enrolment into chemotherapy, which would facilitate individualized targeted therapy for these subgroups of patients to ensure better survival and an improved quality of life and overall outcome. Harnessing the immune response through immunotherapy approaches has changed the treatment way for patients with cancer. The immune outline has emerged as a beneficial tool for recognizing predictive and prognostic biomarkers clinically. Studying the tumour microenvironment (TME) of ovarian cancer tissue may provide awareness of actionable targets for enhancing chemotherapy outcomes and quality of life. This review analyses the relevance of immunohistochemistry biomarkers as prognostic biomarkers in predicting chemotherapy resistance and improving the quality of life in ovarian cancer.
Collapse
|
7
|
Alwosaibai K, Al‑Hujaily E, Alamri S, Ghandorah S, Garson K, Vanderhyden B. PAX2 induces vascular‑like structures in normal ovarian cells and ovarian cancer. Exp Ther Med 2022; 23:412. [PMID: 35601066 PMCID: PMC9117948 DOI: 10.3892/etm.2022.11339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
In adult tissue, the paired box 2 (PAX2) protein is expressed in healthy oviductal, but not normal ovarian surface epithelial cells. PAX2 is expressed in a subset of cases of serous ovarian carcinoma; however, the role of PAX2 in the initiation and progression of ovarian cancer remains unknown. The aim of the present study was to determine the biological effects of PAX2 expression in normal and cancerous epithelial cells. By culturing the normal and cancerous ovarian cells that express PAX2 in 3D culture and staining the cells with vasculogenic mimicry markers such as CD31 and PAS, it was shown that PAX2 overexpression in both normal and cancerous ovarian epithelial cells induced formation of vascular-like structures both in vitro and in vivo. These results indicated a potential role of PAX2 in ovarian cancer progression by increasing the presence of vascular-like structures to promote the supply of nutrients to tumor cells and facilitate cancer cell proliferation and invasion.
Collapse
Affiliation(s)
- Kholoud Alwosaibai
- Biomedical Research Department, Research Center, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia
| | - Ensaf Al‑Hujaily
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Salmah Alamri
- Biomedical Research Department, Research Center, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia
| | - Salim Ghandorah
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia
| | - Kenneth Garson
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Barbara Vanderhyden
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
8
|
Tung LT, Wang H, Belle JI, Petrov JC, Langlais D, Nijnik A. p53-dependent induction of P2X7 on hematopoietic stem and progenitor cells regulates hematopoietic response to genotoxic stress. Cell Death Dis 2021; 12:923. [PMID: 34625535 PMCID: PMC8501024 DOI: 10.1038/s41419-021-04202-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/27/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023]
Abstract
Stem and progenitor cells are the main mediators of tissue renewal and repair, both under homeostatic conditions and in response to physiological stress and injury. Hematopoietic system is responsible for the regeneration of blood and immune cells and is maintained by bone marrow-resident hematopoietic stem and progenitor cells (HSPCs). Hematopoietic system is particularly susceptible to injury in response to genotoxic stress, resulting in the risk of bone marrow failure and secondary malignancies in cancer patients undergoing radiotherapy. Here we analyze the in vivo transcriptional response of HSPCs to genotoxic stress in a mouse whole-body irradiation model and, together with p53 ChIP-Seq and studies in p53-knockout (p53KO) mice, characterize the p53-dependent and p53-independent branches of this transcriptional response. Our work demonstrates the p53-independent induction of inflammatory transcriptional signatures in HSPCs in response to genotoxic stress and identifies multiple novel p53-target genes induced in HSPCs in response to whole-body irradiation. In particular, we establish the direct p53-mediated induction of P2X7 expression on HSCs and HSPCs in response to genotoxic stress. We further demonstrate the role of P2X7 in hematopoietic response to acute genotoxic stress, with P2X7 deficiency significantly extending mouse survival in irradiation-induced hematopoietic failure. We also demonstrate the role of P2X7 in the context of long-term HSC regenerative fitness following sublethal irradiation. Overall our studies provide important insights into the mechanisms of HSC response to genotoxic stress and further suggest P2X7 as a target for pharmacological modulation of HSC fitness and hematopoietic response to genotoxic injury.
Collapse
Affiliation(s)
- Lin Tze Tung
- Department of Physiology, McGill University, Montreal, QC, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - HanChen Wang
- Department of Physiology, McGill University, Montreal, QC, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Jad I Belle
- Department of Physiology, McGill University, Montreal, QC, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Jessica C Petrov
- Department of Physiology, McGill University, Montreal, QC, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - David Langlais
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University Genome Centre, McGill University, Montreal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, QC, Canada.
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada.
| |
Collapse
|
9
|
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
|
10
|
Chui MH, Boroujeni AM, Mandelker D, Ladanyi M, Soslow RA. Characterization of TP53-wildtype tubo-ovarian high-grade serous carcinomas: rare exceptions to the binary classification of ovarian serous carcinoma. Mod Pathol 2021; 34:490-501. [PMID: 32801341 PMCID: PMC8409220 DOI: 10.1038/s41379-020-00648-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 01/04/2023]
Abstract
While TP53 mutation is widely considered to be a defining feature of tubo-ovarian high-grade serous carcinoma (HGSC), rare TP53-mutation-negative cases have been reported. To gain further insight into this rare subset, a retrospective review was conducted on 25 TP53-wildtype tubo-ovarian HGSCs, constituting 2.5% of 987 HGSCs profiled by the MSK-IMPACT sequencing platform. Consistent with serous differentiation, positive staining for Pax8 and WT1 was present in virtually all TP53-wildtype HGSCs. Other characteristic features of HGSC, such as serous tubal intraepithelial carcinoma, or genetic alterations of CCNE1 and BRCA1/2 were identified in these tumors, furthering supporting their classification as bona fide HGSC, despite lacking TP53 mutations. Overall, the level of chromosomal instability of TP53-wildtype HGSCs was intermediate between low-grade serous carcinoma (LGSC) and TP53-mutated HGSC. Morphologic assessment by observers blinded to mutation status revealed a significant subset of tumors with Grade 2 nuclear atypia (which exceeds the degree of atypia allowed for LGSC, but less than typically encountered for HGSC) combined with micropapillary features (6/19, 32%, chemotherapy-naive TP53-wildtype HGSCs compared to 0/21, 0%, TP53-mutated HGSCs; p = 0.007). Some TP53-wildtype HGSCs harbored driver mutations in KRAS (n = 3), BRAF (n = 1) or NRAS (n = 2). Overall, 10 (40%) cases had "LGSC-like" morphology (i.e., Grade 2 nuclear atypia and micropapillary features) and/or RAS/RAF mutation, and most of these showed a wildtype p53 pattern of expression by immunohistochemistry (7/9, 78%). The remaining TP53-wildtype HGSCs (n = 15, 60%) exhibited severe nuclear atypia (Grade 3) and were morphologically indistinguishable from conventional TP53-mutated HGSC. Despite lacking genetic alterations of TP53, these "usual HGSC-like" tumors often showed evidence of p53 dysfunction, including downregulation of expression ('null' or equivocal p53 staining in 9/14, 64%) or MDM2 amplification (n = 2). Our results support the existence of TP53-wildtype HGSCs, which comprise a heterogeneous group of tumors which may arise via distinct pathogenic mechanisms.
Collapse
|
11
|
Izaguirre DI, Ng CW, Kwan SY, Kun EH, Tsang YTM, Gershenson DM, Wong KK. The Role of GDF15 in Regulating the Canonical Pathways of the Tumor Microenvironment in Wild-Type p53 Ovarian Tumor and Its Response to Chemotherapy. Cancers (Basel) 2020; 12:cancers12103043. [PMID: 33086658 PMCID: PMC7650722 DOI: 10.3390/cancers12103043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/10/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Patients with wild-type p53 ovarian cancer appear to have a poorer survival rate than those with mutant p53 due to resistance to chemotherapy. The mechanism underlying this observation is not clearly understood. The aim of this study was to identify potential biomarkers regulated by p53 that conferred resistance using in vitro and in vivo studies. Growth differentiation factor 15 (GDF15) expression was demonstrated to be controlled by p53 in both ovarian cancer cell lines and orthotopic mouse models. The histological and RNAseq studies of the GDF15-knocked down, A2780 cell line-induced tumor revealed that the ratio and canonical pathways of stromal/tumor were modified by secretory GDF15. Abstract Background: The standard treatment of ovarian cancer is surgery followed by a chemotherapeutic combination consisting of a platinum agent, such as cisplatin and a taxane-like paclitaxel. We previously observed that patients with ovarian cancer wild-type for p53 had a poorer survival rate than did those with p53 mutations. Thus, a better understanding of the molecular changes of epithelial ovarian cancer cells with wild-type p53 in response to treatment with cisplatin could reveal novel mechanisms of chemoresistance. Methods: Gene expression profiling was performed on an ovarian cancer cell line A2780 with wild-type p53 treated with cisplatin. A gene encoding a secretory protein growth differentiation factor 15 (GDF15) was identified to be highly induced by cisplatin treatment in vitro. This was further validated in a panel of wild-type and mutant p53 ovarian cancer cell lines, as well as in mouse orthotopic models. The mouse tumor tissues were further analyzed by histology and RNA-seq. Results: GDF15 was identified as one of the highly induced genes by cisplatin or carboplatin in ovarian cancer cell lines with wild-type p53. The wild-type p53-induced expression of GDF15 and GDF15-confered chemotherapy resistance was further demonstrated in vitro and in vivo. This study also discovered that GDF15-knockdown (GDF15-KD) tumors had less stromal component and had different repertoires of activated and inhibited canonical pathways in the stromal cell and cancer cell components from that of the control tumors after cisplatin treatment. Conclusions: GDF15 expression from the wild-type p53 cancer cells can modulate the canonical pathways in the tumor microenvironment in response to cisplatin, which is a possible mechanism of chemoresistance.
Collapse
Affiliation(s)
- Daisy I. Izaguirre
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.I.I.); (C.-W.N.); (S.-Y.K.); (E.H.K.); (Y.T.M.T.); (D.M.G.)
- Cancer Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Chun-Wai Ng
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.I.I.); (C.-W.N.); (S.-Y.K.); (E.H.K.); (Y.T.M.T.); (D.M.G.)
| | - Suet-Yan Kwan
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.I.I.); (C.-W.N.); (S.-Y.K.); (E.H.K.); (Y.T.M.T.); (D.M.G.)
- Cancer Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Eucharist H. Kun
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.I.I.); (C.-W.N.); (S.-Y.K.); (E.H.K.); (Y.T.M.T.); (D.M.G.)
| | - Yvonne T. M. Tsang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.I.I.); (C.-W.N.); (S.-Y.K.); (E.H.K.); (Y.T.M.T.); (D.M.G.)
| | - David M. Gershenson
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.I.I.); (C.-W.N.); (S.-Y.K.); (E.H.K.); (Y.T.M.T.); (D.M.G.)
| | - Kwong-Kwok Wong
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.I.I.); (C.-W.N.); (S.-Y.K.); (E.H.K.); (Y.T.M.T.); (D.M.G.)
- Cancer Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-713-792-0229
| |
Collapse
|
12
|
Voutsadakis IA. Low-grade serous ovarian carcinoma: an evolution toward targeted therapy. Int J Gynecol Cancer 2020; 30:1619-1626. [PMID: 31780569 DOI: 10.1136/ijgc-2019-000832] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/16/2019] [Accepted: 09/24/2019] [Indexed: 11/04/2022] Open
Abstract
Low-grade serous ovarian carcinoma and its high-grade serous ovarian carcinoma counterpart differ in their precursor lesions, molecular profile, natural history, and response to therapies. As such, low-grade serous ovarian carcinoma needs to be studied separately from high-grade serous ovarian carcinoma, despite challenges stemming from its rarity. A deeper understanding of the pathogenesis of low-grade serous ovarian carcinoma and the most common molecular defects and pathways involved in the carcinogenesis of the ovarian epithelium from normal to serous borderline ovarian tumors to low-grade serous ovarian carcinoma will help develop better therapies. By adopting targeted approaches there may be an opportunity to integrate novel therapies without the need for robust numbers in clinical trials. This manuscript will discuss low-grade serous ovarian carcinoma and focus on the arising treatments being developed with an improved understanding of the pathogenesis of this disease.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Section of Internal Medicine Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
13
|
Mota A, S Oltra S, Moreno-Bueno G. Insight updating of the molecular hallmarks in ovarian carcinoma. EJC Suppl 2020; 15:16-26. [PMID: 33240439 PMCID: PMC7573468 DOI: 10.1016/j.ejcsup.2019.11.001] [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: 04/12/2019] [Revised: 10/17/2019] [Accepted: 11/16/2019] [Indexed: 12/31/2022] Open
Abstract
Background and purpose Ovarian cancer (OC) is the deadliest gynaecologic cancer characterised by a high heterogeneity not only at the clinical point of view but also at the molecular level. This review focuses on the new insights about the OC molecular classification. Materials and methods We performed a bibliographic search for different indexed articles focused on the new molecular classification of OC. All of them have been published in PubMed and included information about the most frequent molecular alterations in OC confirmed by omics approaches. In addition, we have extracted information about the role of liquid biopsy in the OC diagnosis and prognosis. Results New molecular insights into OC have allowed novel clinical entities to be defined. Among OC, high-grade serous ovarian carcinoma (HGSOC) which is the most common OC is characterised by omics approaches, mutations in TP53 and in other genes involved in the homologous recombination repair, especially BRCA1/2. Recent studies in HGSOC have allowed a new molecular classification in subgroups according to their mutational, transcriptional, methylation and copy number variation signatures with a real impact in the characterisation of new therapeutic targets for OC to be defined. Furthermore, despite the intrinsic intra-tumour heterogeneity, the advances in next generation sequencing (NGS) analyses of ascetic liquid from OC have opened new ways for its characterisation and treatment. Conclusions The advances in genomic approaches have been used for the identification of new molecular profiling techniques which define OC subgroups and has supposed advances in the diagnosis and in the personalised treatment of OC. Classification of ovarian cancer regarding to widespread genetic and genomic data. Highlighted role of p53 and BRCA1/2 in ovarian cancer for diagnosis and treatment. Intra-tumour genetic heterogeneity in ovarian cancer. Useful of liquid biopsy study in ovarian cancer diagnosis.
Collapse
Affiliation(s)
- Alba Mota
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), IdiPaz, MD Anderson International Foundation Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Sara S Oltra
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), IdiPaz, MD Anderson International Foundation Madrid, Spain
| | - Gema Moreno-Bueno
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), IdiPaz, MD Anderson International Foundation Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
- Corresponding author: Departamento de Bioquímica, Facultad de Medicina (UAM), Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, Arzobispo Morcillo 4, Madrid, 28029, Spain. Fax: +34 91-5854401.
| |
Collapse
|
14
|
Frisone D, Charrier M, Clement S, Christinat Y, Thouvenin L, Homicsko K, Michielin O, Bodmer A, Chappuis PO, McKee TA, Tsantoulis P. Durable response to palbociclib and letrozole in ovarian cancer with CDKN2A loss. Cancer Biol Ther 2019; 21:197-202. [PMID: 31709901 PMCID: PMC7012162 DOI: 10.1080/15384047.2019.1685291] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 10/13/2019] [Accepted: 10/22/2019] [Indexed: 01/27/2023] Open
Abstract
Alterations of the Retinoblastoma (Rb) pathway are frequent in ovarian cancer, typically resulting from CDKN2A down-regulation, CCNE1 amplification, CCND1/2 amplification, and RB1 loss. However, bi-allelic CDKN2A mutation or homozygous deletion is a very rare event, concerning less than 5% of patients.Initial trials with palbociclib in serous ovarian cancer have shown very modest benefit in unselected patient populations, thus underlining the need for a biomarker predicting response. We report the case of a heavily pre-treated patient with a serous ovarian tumor harboring a homozygous deletion of the CDKN2A gene that derived significant, prolonged clinical benefit from palbociclib, a CDK4/6 oral inhibitor, with letrozole. Treatment with palbociclib and letrozole started on February 2018, with an ongoing response after 12 months.In conclusion, homozygous CDKN2A deletion is rare and could be used to predict response to CDK4/6 inhibitors in association with other genomic features. We encourage further trials in this direction.
Collapse
Affiliation(s)
- Daniele Frisone
- Department of Oncology, University Hospitals of Geneva (HUG), Geneva, Switzerland
| | - Melinda Charrier
- Department of Oncology, University Hospitals of Geneva (HUG), Geneva, Switzerland
| | - Sophie Clement
- Department of Genetic Medicine, Laboratory and Pathology, University Hospitals of Geneva (HUG), Geneva, Switzerland
| | - Yann Christinat
- Department of Genetic Medicine, Laboratory and Pathology, University Hospitals of Geneva (HUG), Geneva, Switzerland
| | - Laure Thouvenin
- Department of Oncology, University Hospitals of Geneva (HUG), Geneva, Switzerland
| | - Krisztian Homicsko
- Multidisciplinary Oncology Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Olivier Michielin
- Multidisciplinary Oncology Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Alexandre Bodmer
- Department of Oncology, University Hospitals of Geneva (HUG), Geneva, Switzerland
| | - Pierre O. Chappuis
- Department of Oncology, University Hospitals of Geneva (HUG), Geneva, Switzerland
- Department of Genetic Medicine, Laboratory and Pathology, University Hospitals of Geneva (HUG), Geneva, Switzerland
| | - Thomas A. McKee
- Department of Genetic Medicine, Laboratory and Pathology, University Hospitals of Geneva (HUG), Geneva, Switzerland
| | - Petros Tsantoulis
- Department of Oncology, University Hospitals of Geneva (HUG), Geneva, Switzerland
- Department of Medical Specialties Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
15
|
Influence of p53 Isoform Expression on Survival in High-Grade Serous Ovarian Cancers. Sci Rep 2019; 9:5244. [PMID: 30918304 PMCID: PMC6437169 DOI: 10.1038/s41598-019-41706-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 03/14/2019] [Indexed: 11/08/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is characterised by alterations in the p53 pathway. The expression levels of p53 isoforms have been shown to be associated with patient survival in several cancers. This study examined the predictive and prognostic effects of the expression levels of TP53 pre-mRNA splicing isoforms and TP53 mutations in tumour tissues in 40 chemotherapy responders and 29 non-responders with HGSOC. The mRNA expression levels from total p53, and total Δ133p53, p53β, p53γ isoforms were determined by RT-qPCR, and TP53 mutation status by targeted massive parallel sequencing. The results from these analyses were correlated with the clinical outcome parameters. No differential expression of p53 isoforms could be detected between the chemosensitive and chemoresistant subgroups. In a multivariate Cox regression model, high levels of total Δ133p53 were found to be an independent prognosticator for improved overall survival (HR = 0.422, p = 0.018, 95% CI: 0.207–0.861) and reached borderline significance for progression-free survival (HR = 0.569, p = 0.061, 95% CI: 0.315–1.027). TP53 mutations resulting in loss of function or located at known hotspots were predictive of tumour characteristics and disease progression. These findings suggest that total Δ133p53 mRNA can be a biomarker for survival in HGSOC.
Collapse
|
16
|
Abstract
High-grade serous ovarian carcinoma (HGSOC) is characterised by alterations in the p53 pathway. The expression levels of p53 isoforms have been shown to be associated with patient survival in several cancers. This study examined the predictive and prognostic effects of the expression levels of TP53 pre-mRNA splicing isoforms and TP53 mutations in tumour tissues in 40 chemotherapy responders and 29 non-responders with HGSOC. The mRNA expression levels from total p53, and total Δ133p53, p53β, p53γ isoforms were determined by RT-qPCR, and TP53 mutation status by targeted massive parallel sequencing. The results from these analyses were correlated with the clinical outcome parameters. No differential expression of p53 isoforms could be detected between the chemosensitive and chemoresistant subgroups. In a multivariate Cox regression model, high levels of total Δ133p53 were found to be an independent prognosticator for improved overall survival (HR = 0.422, p = 0.018, 95% CI: 0.207-0.861) and reached borderline significance for progression-free survival (HR = 0.569, p = 0.061, 95% CI: 0.315-1.027). TP53 mutations resulting in loss of function or located at known hotspots were predictive of tumour characteristics and disease progression. These findings suggest that total Δ133p53 mRNA can be a biomarker for survival in HGSOC.
Collapse
|
17
|
Breed C, Hicks DA, Webb PG, Galimanis CE, Bitler BG, Behbakht K, Baumgartner HK. Ovarian Tumor Cell Expression of Claudin-4 Reduces Apoptotic Response to Paclitaxel. Mol Cancer Res 2019; 17:741-750. [PMID: 30606772 DOI: 10.1158/1541-7786.mcr-18-0451] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 10/11/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023]
Abstract
A significant factor contributing to poor survival rates for patients with ovarian cancer is the insensitivity of tumors to standard-of-care chemotherapy. In this study, we investigated the effect of claudin-4 expression on ovarian tumor cell apoptotic response to cisplatin and paclitaxel. We manipulated claudin-4 gene expression by silencing expression [short hairpin RNA (shRNA)] in cells with endogenously expressed claudin-4 or overexpressing claudin-4 in cells that natively do not express claudin-4. In addition, we inhibited claudin-4 activity with a claudin mimic peptide (CMP). We monitored apoptotic response by caspase-3 and Annexin V binding. We examined proliferation rate by counting the cell number over time as well as measuring the number of mitotic cells. Proximity ligation assays, immunoprecipitation (IP), and immunofluorescence were performed to examine interactions of claudin-4. Western blot analysis of tubulin in cell fractions was used to determine the changes in tubulin polymerization with changes in claudin-4 expression. Results show that claudin-4 expression reduced epithelial ovarian cancer (EOC) cell apoptotic response to paclitaxel. EOCs without claudin-4 proliferated more slowly with enhanced mitotic arrest compared with the cells expressing claudin-4. Furthermore, our results indicate that claudin-4 interacts with tubulin, having a profound effect on the structure and polymerization of the microtubule network. In conclusion, we demonstrate that claudin-4 reduces the ovarian tumor cell response to microtubule-targeting paclitaxel and disrupting claudin-4 with CMP can restore apoptotic response. IMPLICATIONS: These results suggest that claudin-4 expression may provide a biomarker for paclitaxel response and can be a target for new therapeutic strategies to improve response.
Collapse
Affiliation(s)
- Christopher Breed
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado.,Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado
| | - Douglas A Hicks
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado
| | - Patricia G Webb
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado
| | - Carly E Galimanis
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado
| | - Benjamin G Bitler
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado
| | - Kian Behbakht
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado.,Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado
| | - Heidi K Baumgartner
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado. .,Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
18
|
Tonnessen-Murray C, Ungerleider NA, Rao SG, Wasylishen AR, Frey WD, Jackson JG. p53 Mediates Vast Gene Expression Changes That Contribute to Poor Chemotherapeutic Response in a Mouse Model of Breast Cancer. Transl Oncol 2018; 11:930-940. [PMID: 29852458 PMCID: PMC6041561 DOI: 10.1016/j.tranon.2018.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 05/03/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
p53 is a transcription factor that regulates expression of genes involved in cell cycle arrest, senescence, and apoptosis. TP53 harbors mutations that inactivate its transcriptional activity in roughly 30% of breast cancers, and these tumors are much more likely to undergo a pathological complete response to chemotherapy. Thus, the gene expression program activated by wild-type p53 contributes to a poor response. We used an in vivo genetic model system to comprehensively define the p53- and p21-dependent genes and pathways modulated in tumors following doxorubicin treatment. We identified genes differentially expressed in spontaneous mammary tumors harvested from treated MMTV-Wnt1 mice that respond poorly (Trp53+/+) or favorably (Trp53-null) and those that lack the critical senescence/arrest p53 target gene Cdkn1a. Trp53 wild-type tumors differentially expressed nearly 10-fold more genes than Trp53-null tumors after treatment. Pathway analyses showed that genes involved in cell cycle, senescence, and inflammation were enriched in treated Trp53 wild-type tumors; however, no genes/pathways were identified that adequately explain the superior cell death/tumor regression observed in Trp53-null tumors. Cdkn1a-null tumors that retained arrest capacity (responded poorly) and those that proliferated (responded well) after treatment had remarkably different gene regulation. For instance, Cdkn1a-null tumors that arrested upregulated Cdkn2a (p16), suggesting an alternative, p21-independent route to arrest. Live animal imaging of longitudinal gene expression of a senescence/inflammation gene reporter in Trp53+/+ tumors showed induction during and after chemotherapy treatment, while tumors were arrested, but expression rapidly diminished immediately upon relapse.
Collapse
Affiliation(s)
- Crystal Tonnessen-Murray
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, 70112
| | | | - Sonia G Rao
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, 70112
| | - Amanda R Wasylishen
- Department of Genetics, MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Wesley D Frey
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, 70112
| | - James G Jackson
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, 70112.
| |
Collapse
|
19
|
Martin EG, Law J, Ran W, Helbig N, Birkhead GS. Evaluating the Quality and Usability of Open Data for Public Health Research: A Systematic Review of Data Offerings on 3 Open Data Platforms. JOURNAL OF PUBLIC HEALTH MANAGEMENT AND PRACTICE 2018; 23:e5-e13. [PMID: 26910872 DOI: 10.1097/phh.0000000000000388] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
CONTEXT Government datasets are newly available on open data platforms that are publicly accessible, available in nonproprietary formats, free of charge, and with unlimited use and distribution rights. They provide opportunities for health research, but their quality and usability are unknown. OBJECTIVE To describe available open health data, identify whether data are presented in a way that is aligned with best practices and usable for researchers, and examine differences across platforms. DESIGN Two reviewers systematically reviewed a random sample of data offerings on NYC OpenData (New York City, all offerings, n = 37), Health Data NY (New York State, 25% sample, n = 71), and HealthData.gov (US Department of Health and Human Services, 5% sample, n = 75), using a standard coding guide. SETTING Three open health data platforms at the federal, New York State, and New York City levels. MAIN OUTCOME MEASURES Data characteristics from the coding guide were aggregated into summary indices for intrinsic data quality, contextual data quality, adherence to the Dublin Core metadata standards, and the 5-star open data deployment scheme. RESULTS One quarter of the offerings were structured datasets; other presentation styles included charts (14.7%), documents describing data (12.0%), maps (10.9%), and query tools (7.7%). Health Data NY had higher intrinsic data quality (P < .001), contextual data quality (P < .001), and Dublin Core metadata standards adherence (P < .001). All met basic "web availability" open data standards; fewer met higher standards of "hyperlinked to other data." CONCLUSIONS Although all platforms need improvement, they already provide readily available data for health research. Sustained effort on improving open data websites and metadata is necessary for ensuring researchers use these data, thereby increasing their research value.
Collapse
Affiliation(s)
- Erika G Martin
- Nelson A. Rockefeller Institute of Government, Albany, New York (Dr Martin); Rockefeller College of Public Affairs & Policy (Dr Martin and Ms Law), College of Computing and Information (Ms Ran), and School of Public Health (Dr Birkhead), University at Albany, Albany, New York; and New York State Department of Health, Albany, New York, NY (Drs Helbig and Birkhead)
| | | | | | | | | |
Collapse
|
20
|
Zhu S, Chen Z, Wang L, Peng D, Belkhiri A, Lockhart AC, El-Rifai W. A Combination of SAHA and Quinacrine Is Effective in Inducing Cancer Cell Death in Upper Gastrointestinal Cancers. Clin Cancer Res 2018; 24:1905-1916. [PMID: 29386219 DOI: 10.1158/1078-0432.ccr-17-1716] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 10/27/2017] [Accepted: 01/25/2018] [Indexed: 11/16/2022]
Abstract
Purpose: We aimed to investigate the therapeutic efficacy of single agent and the combination of quinacrine and suberoylanilide hydroxamic acid (SAHA) in wt- and mut-p53 upper gastrointestinal cancer (UGC) cell models.Experimental Design: ATP-Glo, clonogenic cell survival, Annexin V, comet, DNA double-strand breaks (DSBs), qPCR, and Western blot analysis assays were utilized.Results: Using clonogenic cell survival, ATP-Glo cell viability, Annexin V, and sub-G0 population analysis, we demonstrated that a combination of quinacrine and SAHA significantly decreased colony formation and increased cancer cell death (range, 4-20 fold) in six UGC cell models, as compared with single-agent treatments, irrespective of the p53 status (P < 0.01). The combination of quinacrine and SAHA induced high levels of DSB DNA damage (>20-fold, P < 0.01). Western blot analysis showed activation of caspases-3, 9, and γ-H2AX in all cell models. Of note, although quinacrine treatment induced expression of wt-p53 protein, the combination of quinacrine and SAHA substantially decreased the levels of both wt-P53 and mut-P53. Furthermore, cell models that were resistant to cisplatin (CDDP) or gefitinib treatments were sensitive to this combination. Tumor xenograft data confirmed that a combination of quinacrine and SAHA is more effective than a single-agent treatment in abrogating tumor growth in vivo (P < 0.01).Conclusions: Our novel findings show that the combination of quinacrine and SAHA promotes DNA damage and is effective in inducing cancer cell death, irrespective of p53 status and resistance to CDDP or gefitinib in UGC models. Clin Cancer Res; 24(8); 1905-16. ©2018 AACR.
Collapse
Affiliation(s)
- Shoumin Zhu
- Department of Surgery, University of Miami, Miami, Florida
| | - Zheng Chen
- Department of Surgery, University of Miami, Miami, Florida
| | - Lihong Wang
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dunfa Peng
- Department of Surgery, University of Miami, Miami, Florida
| | - Abbes Belkhiri
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - A Craig Lockhart
- Division of Medical Oncology, University of Miami, Miami, Florida
| | - Wael El-Rifai
- Department of Surgery, University of Miami, Miami, Florida. .,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida.,Department of Veterans Affairs, Miami Healthcare System, Miami, Florida
| |
Collapse
|
21
|
Bhatt M, Ivan C, Xie X, Siddik ZH. Drug-dependent functionalization of wild-type and mutant p53 in cisplatin-resistant human ovarian tumor cells. Oncotarget 2017; 8:10905-10918. [PMID: 28038466 PMCID: PMC5355233 DOI: 10.18632/oncotarget.14228] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 12/18/2016] [Indexed: 01/12/2023] Open
Abstract
Cisplatin (cis-Pt) resistance in tumor cells from p53 dysfunction is a significant clinical problem. Although mutation can inhibit p53 function, >60% of p53 mutants retain normal function according to literature reports. Therefore, we examined the status of p53 in cisplatin-resistant ovarian tumor models and its functional response to cis-Pt and the mechanistically-distinct non-cross-resistant oxaliplatin (oxali-Pt). Relative to sensitive A2780 cells harboring wild-type p53, the 2780CP/Cl-16, OVCAR-10, Hey and OVCA-433 cell lines were 10- to 30-fold resistant to cis-Pt, but was substantially circumvented by oxali-Pt. Mutant p53 in 2780CP/Cl-16 (p53V172F) and OVCAR-10 (p53V172F and p53G266R) cells, predicted as non-functional in p53 database, displayed attenuated response to cis-Pt, as did the polymorphic p53P72R (functionally equivalent to wild-type p53) in HEY and OVCA-433 cell lines. However, p53 was robustly activated by oxali-Pt in all cell lines, with resultant drug potency confirmed as p53-dependent by p53 knockout using CRISPR/Cas9 system. This p53 activation by oxali-Pt was associated with phosphorylation at Ser20 by MEK1/2 based on inhibitor and kinase studies. Cis-Pt, however, failed to phosphorylate Ser20 due to downregulated Chk2, and its clinical impact validated by reduced overall survival of ovarian cancer patients according to TCGA database. In conclusion, cis-Pt resistance occurs in both wild-type and mutant p53 ovarian cancer cells, but is associated with loss of Ser20 phosphorylation. However, these mutant p53, like polymorphic p53, are functional and activated by oxali-Pt-induced Ser20 phosphorylation. Thus, the potential exists for repurposing oxali-Pt or similar drugs against refractory cancers harboring wild-type or specific mutant p53.
Collapse
Affiliation(s)
- Michelle Bhatt
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xiaolei Xie
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zahid H Siddik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
22
|
Shi C, Zhang Z. Screening of potentially crucial genes and regulatory factors involved in epithelial ovarian cancer using microarray analysis. Oncol Lett 2017; 14:725-732. [PMID: 28693226 PMCID: PMC5494615 DOI: 10.3892/ol.2017.6183] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/17/2017] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to screen potential genes implicated in epithelial ovarian cancer (EOC) and to further understand the molecular pathogenesis of EOC. In order to do this, datasets GSE14407 (containing 12 human ovarian cancer epithelia samples and 12 normal epithelia samples) and GSE29220 (containing 11 salivary transcriptomes from ovarian cancer patients with serous papillary adenocarcinoma and 11 matched controls) were obtained from the Gene Expression Omnibus. Differentially expressed genes (DEGs) within these datasets were screened using the Linear Models for Microarray Data package, and potential gene functions were predicted by functional and pathway enrichment analyses. Additionally, module analysis of protein-protein interaction networks was performed using MCODE software in Cytoscape. The potential microRNAs (miRNAs/miRs) and transcription factors (TFs) regulating DEGs were also analyzed, and the integrated TF-DEG and miRNA-DEG regulatory networks were visualized with Cytoscape. In total, 31 upregulated DEGs and 64 downregulated DEGs were screened. The upregulated DEGs, such as centromere protein F (CENPF) and ubiquitin like with PHD and ring finger domains 1 (UHRF1), were significantly associated with the cell cycle and were regulated by the TF nuclear transcription factor Y (NF-Y). CENPF was modulated by miR-373, and UHRF1 was regulated by miR-146a. The downregulated DEGs, such as aldehyde dehydrogenase 1 family member A2 (ALDH1A2), were distinctly involved in the response to estrogen stimulus and modulated by tumor protein 53 (TP53); protocadherin 9 (PCDH9) was regulated by TP53, miR-92b-3p and miR-137. The DEGs, including CENPF, UHRF1, ALDH1A2 and PCDH9, and a set of gene regulators, including all NFY genes, TP53, miR-373, miR-146a, miR-92b-3p and miR-137, may be involved in the pathogenesis of EOC.
Collapse
Affiliation(s)
- Can Shi
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Zhenyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| |
Collapse
|
23
|
Molecular Alterations of TP53 are a Defining Feature of Ovarian High-Grade Serous Carcinoma: A Rereview of Cases Lacking TP53 Mutations in The Cancer Genome Atlas Ovarian Study. Int J Gynecol Pathol 2017; 35:48-55. [PMID: 26166714 DOI: 10.1097/pgp.0000000000000207] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The Cancer Genome Atlas has reported that 96% of ovarian high-grade serous carcinomas (HGSCs) have TP53 somatic mutations suggesting that mutation of this gene is a defining feature of this neoplasm. In the current study, 5 gynecologic pathologists independently evaluated hematoxylin and eosin slides of 14 available cases from The Cancer Genome Atlas classified as HGSC that lacked a TP53 mutation. The histologic diagnoses rendered by these pathologists and the accompanying molecular genetic data are the subject of this report. Only 1 case (Case 5), which contained a homozygous deletion of TP53, had unanimous interobserver agreement for a diagnosis of pure HGSC. In 1 case (Case 3), all 5 observers (100%) rendered a diagnosis of HGSC; however, 3 observers (60%) noted that the histologic features were not classic for HGSC and suggested this case may have arisen from a low-grade serous carcinoma (arisen from an alternate pathway compared with the usual HGSC). In 2 cases (Cases 4 and 12), only 3 observers (60%) in each case, respectively, interpreted it as having a component of HGSC. In the remaining 10 (71%) of tumors (Cases 1, 2, 6-11, 13, and 14), the consensus diagnosis was not HGSC, with individual diagnoses including low-grade serous carcinoma, high-grade endometrioid carcinoma, HGSC, metastatic carcinoma, clear cell carcinoma, atypical proliferative (borderline) serous tumor, and adenocarcinoma, not otherwise specified. Therefore, 13 (93%) of the tumors (Cases 1-4 and 6-14) were either not a pure HGSC or represented a diagnosis other than HGSC, all with molecular results not characteristic of HGSC. Accordingly, our review of the TP53 wild-type HGSCs reported in The Cancer Genome Atlas suggests that 100% of de novo HGSCs contain TP53 somatic mutations or deletions, with the exception of the rare HGSCs that develop from a low-grade serous tumor precursor. We, therefore, propose that lack of molecular alterations of TP53 are essentially inconsistent with the diagnosis of ovarian HGSC and that tumors diagnosed as such should be rigorously reassessed to achieve correct classification.
Collapse
|
24
|
Sun X, Wang X, Feng W, Guo H, Tang C, Lu Y, Xiang X, Bao Y. Gene signatures associated with drug resistance to irinotecan and oxaliplatin predict a poor prognosis in patients with colorectal cancer. Oncol Lett 2017; 13:2089-2096. [PMID: 28454366 PMCID: PMC5403337 DOI: 10.3892/ol.2017.5691] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 11/30/2016] [Indexed: 12/15/2022] Open
Abstract
The identification of novel survival predictors may help to improve the appropriate management of colorectal cancer (CRC). In the present study, two gene sets associated with irinotecan or oxaliplatin resistance in CRC cell lines were first identified and subsequently applied to the clinical CRC microarray dataset GSE14333. Subsequently, a 60-gene irinotecan resistance-associated signature and a 13-gene oxaliplatin resistance-associated signature were established, which were able to classify CRC patients into high- and low-risk subgroups with varied clinical outcomes [irinotecan-resistance gene signature: hazard ratio (HR)=0.4607, 95% confidence interval (CI)=0.3369-0.6300, P<0.0001; oxaliplatin-resistance gene signature: HR=0.6119, 95% CI=0.4547-0.8233, P=0.0008]. The performance of these two gene expression signatures in predicting outcome risk were also validated in two other independent CRC gene expression microarray datasets, GSE17536 (irinotecan-resistance gene signature: HR=0.5318, 95% CI=0.3359-0.8419, P=0.0079; oxaliplatin-resistance gene signature: HR=0.5383, 95% CI=0.3400-0.8521, P=0.0114) and GSE17537 (irinotecan-resistance gene signature: HR=0.2827, 95% CI=0.1173-0.6813, P=0.0088; oxaliplatin-resistance gene signature: HR=0.2378, 95% CI=0.09773-0.5784, P=0.0023). Furthermore, the combination of these two gene classifiers demonstrated a superior performance in CRC prognosis prediction than either used individually. Therefore, this study proposed novel gene classifier models for CRC prognosis prediction, which may be potentially useful to inform treatment decisions for patients with CRC in clinical settings.
Collapse
Affiliation(s)
- Xinrong Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Huzhou Teachers College, The First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, P.R. China
| | - Xiang Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Huzhou Teachers College, The First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, P.R. China
| | - Wenming Feng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Huzhou Teachers College, The First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, P.R. China
| | - Huihui Guo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Huzhou Teachers College, The First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, P.R. China
| | - Chengwu Tang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Huzhou Teachers College, The First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, P.R. China
| | - Yongliang Lu
- Department of Medicine, Huzhou Teachers College, Huzhou, Zhejiang 313000, P.R. China
| | - Xiaobin Xiang
- Department of Sports Medicine, Guangdong Orthopedic Hospital, Guangzhou, Guangdong 510045, P.R. China
| | - Ying Bao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Huzhou Teachers College, The First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, P.R. China
| |
Collapse
|
25
|
Tonnessen-Murray CA, Lozano G, Jackson JG. The Regulation of Cellular Functions by the p53 Protein: Cellular Senescence. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026112. [PMID: 27881444 DOI: 10.1101/cshperspect.a026112] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Transformed cells have properties that allow them to survive and proliferate inappropriately. These characteristics often arise as a result of mutations caused by DNA damage. p53 suppresses transformation by removing the proliferative or survival capacity of cells with DNA damage or inappropriate cell-cycle progression. Cellular senescence, marked by morphological and gene expression changes, is a critical component of p53-mediated tumor suppression. In response to stress, p53 can facilitate an arrest and senescence program in cells exposed to stresses such as DNA damage and oncogene activation, preventing transformation. Senescent cells are evident in precancerous adenoma-type lesions, whereas proliferating, malignant tumors have bypassed senescence, either by p53 mutation or inactivation of the p53 pathway by other means. Tumors that have retained wild-type p53 often show a p53-mediated senescence response to chemotherapy. This response is actually detrimental in some tumor types, as senescent cells can drive relapse by persisting and producing cytokines and chemokines through an acquired secretory phenotype.
Collapse
Affiliation(s)
- Crystal A Tonnessen-Murray
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, Louisiana 70112
| | - Guillermina Lozano
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - James G Jackson
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, Louisiana 70112
| |
Collapse
|
26
|
SASP: Tumor Suppressor or Promoter? Yes! Trends Cancer 2016; 2:676-687. [PMID: 28741506 DOI: 10.1016/j.trecan.2016.10.001] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 09/27/2016] [Accepted: 10/04/2016] [Indexed: 01/07/2023]
Abstract
Cellular senescence is a permanent growth arrest in cells with damage or stress that could lead to transformation. Some tumor cells also undergo senescence in response to chemotherapy. Senescent cells secrete cytokines and other factors of the senescence-associated secretory phenotype (SASP) that contribute to tumor suppression by enforcing arrest and recruiting immune cells that remove these damaged or oncogene-expressing cells from organisms. However, some cells can develop a SASP comprising factors that are immunosuppressive and protumorigenic by paracrine mechanisms. Likewise, the SASP in treated cancers can either contribute to durable responses or drive relapse. Here, we discuss the studies that have demonstrated a complex and often conflicting role for the SASP in tumorigenesis and treatment response.
Collapse
|
27
|
Molecular profiling of low grade serous ovarian tumours identifies novel candidate driver genes. Oncotarget 2016; 6:37663-77. [PMID: 26506417 PMCID: PMC4741956 DOI: 10.18632/oncotarget.5438] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 08/22/2015] [Indexed: 12/31/2022] Open
Abstract
Low grade serous ovarian tumours are a rare and under-characterised histological subtype of epithelial ovarian tumours, with little known of the molecular drivers and facilitators of tumorigenesis beyond classic oncogenic RAS/RAF mutations. With a move towards targeted therapies due to the chemoresistant nature of this subtype, it is pertinent to more fully characterise the genetic events driving this tumour type, some of which may influence response to therapy and/or development of drug resistance. We performed genome-wide high-resolution genomic copy number analysis (Affymetrix SNP6.0) and mutation hotspot screening (KRAS, BRAF, NRAS, HRAS, ERBB2 and TP53) to compare a large cohort of ovarian serous borderline tumours (SBTs, n = 57) with low grade serous carcinomas (LGSCs, n = 19). Whole exome sequencing was performed for 13 SBTs, nine LGSCs and one mixed low/high grade carcinoma. Copy number aberrations were detected in 61% (35/57) of SBTs, compared to 100% (19/19) of LGSCs. Oncogenic RAS/RAF/ERBB2 mutations were detected in 82.5% (47/57) of SBTs compared to 63% (12/19) of LGSCs, with NRAS mutations detected only in LGSC. Some copy number aberrations appeared to be enriched in LGSC, most significantly loss of 9p and homozygous deletions of the CDKN2A/2B locus. Exome sequencing identified BRAF, KRAS, NRAS, USP9X and EIF1AX as the most frequently mutated genes. We have identified markers of progression from borderline to LGSC and novel drivers of LGSC. USP9X and EIF1AX have both been linked to regulation of mTOR, suggesting that mTOR inhibitors may be a key companion treatment for targeted therapy trials of MEK and RAF inhibitors.
Collapse
|
28
|
Specific TP53 Mutants Overrepresented in Ovarian Cancer Impact CNV, TP53 Activity, Responses to Nutlin-3a, and Cell Survival. Neoplasia 2016; 17:789-803. [PMID: 26585234 PMCID: PMC4656807 DOI: 10.1016/j.neo.2015.10.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/19/2015] [Accepted: 10/23/2015] [Indexed: 01/22/2023] Open
Abstract
Evolutionary Action analyses of The Cancer Gene Atlas data sets show that many specific p53 missense and gain-of-function mutations are selectively overrepresented and functional in high-grade serous ovarian cancer (HGSC). As homozygous alleles, p53 mutants are differentially associated with specific loss of heterozygosity (R273; chromosome 17); copy number variation (R175H; chromosome 9); and up-stream, cancer-related regulatory pathways. The expression of immune-related cytokines was selectively related to p53 status, showing for the first time that specific p53 mutants impact, and are related to, the immune subtype of ovarian cancer. Although the majority (31%) of HGSCs exhibit loss of heterozygosity, a significant number (24%) maintain a wild-type (WT) allele and represent another HGSC subtype that is not well defined. Using human and mouse cell lines, we show that specific p53 mutants differentially alter endogenous WT p53 activity; target gene expression; and responses to nutlin-3a, a small molecular that activates WT p53 leading to apoptosis, providing "proof of principle" that ovarian cancer cells expressing WT and mutant alleles represent a distinct ovarian cancer subtype. We also show that siRNA knock down of endogenous p53 in cells expressing homozygous mutant alleles causes apoptosis, whereas cells expressing WT p53 (or are heterozygous for WT and mutant p53 alleles) are highly resistant. Therefore, despite different gene regulatory pathways associated with specific p53 mutants, silencing mutant p53 might be a suitable, powerful, global strategy for blocking ovarian cancer growth in those tumors that rely on mutant p53 functions for survival. Knowing p53 mutational status in HGSC should permit new strategies tailored to control this disease.
Collapse
|
29
|
Seagle BLL, Eng KH, Dandapani M, Yeh JY, Odunsi K, Shahabi S. Survival of patients with structurally-grouped TP53 mutations in ovarian and breast cancers. Oncotarget 2016. [PMID: 26215675 PMCID: PMC4621916 DOI: 10.18632/oncotarget.4080] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The objective of this study was to determine if ovarian cancer patients with a TP53 mutation grouped by location of the mutation within the p53 protein structure exhibit differential survival outcomes. Data from patients with high grade serous ovarian cancer (HGS OvCa) (N = 316) or breast cancer (BrCa) (N = 981) sequenced by The Cancer Genome Atlas (TCGA) was studied by Kaplan-Meier and Cox proportional hazards survival analysis. A TP53 DNA binding domain (BD) missense mutation (MM) occurred in 58.5% (185/316) of HGS OvCas and 16.8% (165/981) of BrCas. Patients with a TP53 DNA BD MM grouped by structural location had significantly different overall survival (OS) and progression free survival (PFS). Median OS (months) of HGS OvCa patients by structural group were: Sheet-loop-helix stabilizers, 31.1; DNA minor groove residue R248, 33.6; Wild-type, 34.2; all other MMs, 44.5; DNA major groove residues, 84.1, and zinc ion coordinating residues, 87.0 (log-rank p = 0.006). PFS of DNA major groove MM cases was longer than TP53 wild-type cases (19.1 versus 10.1 months, log-rank p = 0.038). HGS OvCa and BrCa patients with structurally-grouped TP53 DNA BD MMs have different survival outcomes.
Collapse
Affiliation(s)
- Brandon-Luke L Seagle
- Department of Obstetrics, Gynecology and Reproductive Sciences, Western Connecticut Health Network, Danbury, CT, USA
| | - Kevin H Eng
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Monica Dandapani
- Department of Obstetrics, Gynecology and Reproductive Sciences, Western Connecticut Health Network, Danbury, CT, USA
| | - Judy Y Yeh
- Department of Obstetrics, Gynecology and Reproductive Sciences, Western Connecticut Health Network, Danbury, CT, USA
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Shohreh Shahabi
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Prentice Women's Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
30
|
Mota A, Triviño JC, Rojo-Sebastian A, Martínez-Ramírez Á, Chiva L, González-Martín A, Garcia JF, Garcia-Sanz P, Moreno-Bueno G. Intra-tumor heterogeneity in TP53 null High Grade Serous Ovarian Carcinoma progression. BMC Cancer 2015; 15:940. [PMID: 26620706 PMCID: PMC4666042 DOI: 10.1186/s12885-015-1952-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 11/23/2015] [Indexed: 12/20/2022] Open
Abstract
Background High grade serous ovarian cancer is characterised by high initial response to chemotherapy but poor outcome in the long term due to acquired resistance. One of the main genetic features of this disease is TP53 mutation. The majority of TP53 mutated tumors harbor missense mutations in this gene, correlated with p53 accumulation. TP53 null tumors constitute a specific subgroup characterised by nonsense, frameshift or splice-site mutations associated to complete absence of p53 expression. Different studies show that this kind of tumors may have a worse prognosis than other TP53 mutated HGSC. Methods In this study, we sought to characterise the intra-tumor heterogeneity of a TP53 null HGSC consisting of six primary tumor samples, two intra-pelvic and four extra-pelvic recurrences using exome sequencing and comparative genome hybridisation. Results Significant heterogeneity was found among the different tumor samples, both at the mutational and copy number levels. Exome sequencing identified 102 variants, of which only 42 were common to all three samples; whereas 7 of the 18 copy number changes found by CGH analysis were presented in all samples. Sanger validation of 20 variants found by exome sequencing in additional regions of the primary tumor and the recurrence allowed us to establish a sequence of the tumor clonal evolution, identifying those populations that most likely gave rise to recurrences and genes potentially involved in this process, like GPNMB and TFDP1. Using functional annotation and network analysis, we identified those biological functions most significantly altered in this tumor. Remarkably, unexpected functions such as microtubule-based movement and lipid metabolism emerged as important for tumor development and progression, suggesting its potential interest as therapeutic targets. Conclusions Altogether, our results shed light on the clonal evolution of the distinct tumor regions identifying the most aggressive subpopulations and at least some of the genes that may be implicated in its progression and recurrence, and highlights the importance of considering intra-tumor heterogeneity when carrying out genetic and genomic studies, especially when these are aimed to diagnostic procedures or to uncover possible therapeutic strategies. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1952-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alba Mota
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain. .,MD Anderson International Foundation, Madrid, Spain.
| | | | | | | | - Luis Chiva
- Department of Gynecologic Oncology, MD Anderson Cancer Center, Madrid, Spain.
| | | | - Juan F Garcia
- MD Anderson International Foundation, Madrid, Spain. .,Department of Pathology, MD Anderson Cancer Center, Madrid, Spain.
| | - Pablo Garcia-Sanz
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain. .,MD Anderson International Foundation, Madrid, Spain.
| | - Gema Moreno-Bueno
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain. .,MD Anderson International Foundation, Madrid, Spain.
| |
Collapse
|
31
|
Arsenic R, Braicu EI, Letsch A, Dietel M, Sehouli J, Keilholz U, Ochsenreither S. Cancer-testis antigen cyclin A1 is broadly expressed in ovarian cancer and is associated with prolonged time to tumor progression after platinum-based therapy. BMC Cancer 2015; 15:784. [PMID: 26499264 PMCID: PMC4619521 DOI: 10.1186/s12885-015-1824-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 10/16/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Cyclin A1 is essential for male gametopoiesis. In acute myeloid leukemia, it acts as a leukemia-associated antigen. Cyclin A1 expression has been reported in several epithelial malignancies, including testicular, endometrial, and epithelial ovarian cancer (EOC). We analyzed Cyclin A1 expression in EOC and its correlation with clinical features to evaluate Cyclin A1 as a T-cell target in EOC. METHODS Cyclin A1 mRNA expression in EOC and healthy tissues was quantified by microarray analysis and quantitative real-time PCR (qRT-PCR). Protein expression in clinical samples was assessed by immunohistochemistry (IHC) and was correlated to clinical features. RESULTS Cyclin A1 protein was homogeneously expressed in 43 of 62 grade 3 tumor samples and in 1 of 10 grade 2 specimens (p < 0.001). Survival analysis showed longer time to progression (TTP) among patients with at least moderate Cyclin A1 expression (univariate: p = 0.018, multivariate: p = 0.035). FIGO stage, grading, age, macroscopic residual tumor after debulking, and peritoneal carcinomatosis / distant metastasis had no impact on TTP or overall survival (OS). CONCLUSION Cyclin A1 is highly expressed in most EOCs. The mechanism behind the prolonged TTP in patients with high Cyclin A1 expression warrants further investigation. The frequent, selectively high expression of Cyclin A1 in EOC makes it a promising target for T-cell therapies.
Collapse
Affiliation(s)
- Ruza Arsenic
- Department of Pathology, Institute of Pathology, Charité - University Hospital Berlin, 10117, Berlin, Germany.
| | - Elena Ilona Braicu
- Departement of Gynecology, University Hospital Berlin, 13353, Berlin, Germany.
| | - Anne Letsch
- Department of Hematology, Oncology and Tumor Immunology - University Hospital Berlin, 12200, Berlin, Germany.
| | - Manfred Dietel
- Department of Pathology, Institute of Pathology, Charité - University Hospital Berlin, 10117, Berlin, Germany.
| | - Jalid Sehouli
- Departement of Gynecology, University Hospital Berlin, 13353, Berlin, Germany.
| | - Ulrich Keilholz
- Charité Cancer Comprehensive Center, Charité, 10117, Berlin, Germany.
| | - Sebastian Ochsenreither
- Department of Hematology, Oncology and Tumor Immunology - University Hospital Berlin, 12200, Berlin, Germany.
| |
Collapse
|
32
|
Crane EK, Kwan SY, Izaguirre DI, Tsang YTM, Mullany LK, Zu Z, Richards JS, Gershenson DM, Wong KK. Nutlin-3a: A Potential Therapeutic Opportunity for TP53 Wild-Type Ovarian Carcinomas. PLoS One 2015; 10:e0135101. [PMID: 26248031 PMCID: PMC4527847 DOI: 10.1371/journal.pone.0135101] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/16/2015] [Indexed: 11/20/2022] Open
Abstract
Epithelial ovarian cancer is a diverse molecular and clinical disease, yet standard treatment is the same for all subtypes. TP53 mutations represent a node of divergence in epithelial ovarian cancer histologic subtypes and may represent a therapeutic opportunity in subtypes expressing wild type, including most low-grade ovarian serous carcinomas, ovarian clear cell carcinomas and ovarian endometrioid carcinomas, which represent approximately 25% of all epithelial ovarian cancer. We therefore sought to investigate Nutlin-3a--a therapeutic which inhibits MDM2, activates wild-type p53, and induces apoptosis--as a therapeutic compound for TP53 wild-type ovarian carcinomas. Fifteen epithelial ovarian cancer cell lines of varying histologic subtypes were treated with Nutlin-3a with determination of IC50 values. Western Blot (WB) and quantitative real-time polymerase chain reaction (qRT-PCR) analyses quantified MDM2, p53, and p21 expression after Nutlin-3a treatment. DNA from 15 cell lines was then sequenced for TP53 mutations in exons 2-11 including intron-exon boundaries. Responses to Nutlin-3a were dependent upon TP53 mutation status. By qRT-PCR and WB, levels of MDM2 and p21 were upregulated in wild-type TP53 sensitive cell lines, and p21 induction was reduced or absent in mutant cell lines. Annexin V assays demonstrated apoptosis in sensitive cell lines treated with Nutlin-3a. Thus, Nutlin-3a could be a potential therapeutic agent for ovarian carcinomas expressing wild-type TP53 and warrants further investigation.
Collapse
Affiliation(s)
- Erin K. Crane
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Suet-Yan Kwan
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Cancer Biology Program, The University of Texas at Houston Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Daisy I. Izaguirre
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Cancer Biology Program, The University of Texas at Houston Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Yvonne T. M. Tsang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Lisa K. Mullany
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Zhifei Zu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - JoAnne S. Richards
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - David M. Gershenson
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Kwong-Kwok Wong
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Cancer Biology Program, The University of Texas at Houston Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| |
Collapse
|
33
|
Jeong HH, Leem S, Wee K, Sohn KA. Integrative network analysis for survival-associated gene-gene interactions across multiple genomic profiles in ovarian cancer. J Ovarian Res 2015; 8:42. [PMID: 26138921 PMCID: PMC4491426 DOI: 10.1186/s13048-015-0171-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 06/24/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Recent advances in high-throughput technology and the emergence of large-scale genomic datasets have enabled detection of genomic features that affect clinical outcomes. Although many previous computational studies have analysed the effect of each single gene or the additive effects of multiple genes on the clinical outcome, less attention has been devoted to the identification of gene-gene interactions of general type that are associated with the clinical outcome. Moreover, the integration of information from multiple molecular profiles adds another challenge to this problem. Recently, network-based approaches have gained huge popularity. However, previous network construction methods have been more concerned with the relationship between features only, rather than the effect of feature interactions on clinical outcome. METHODS We propose a mutual information-based integrative network analysis framework (MINA) that identifies gene pairs associated with clinical outcome and systematically analyses the resulting networks over multiple genomic profiles. We implement an efficient non-parametric testing scheme that ensures the significance of detected gene interactions. We develop a tool named MINA that automates the proposed analysis scheme of identifying outcome-associated gene interactions and generating various networks from those interacting pairs for downstream analysis. RESULTS We demonstrate the proposed framework using real data from ovarian cancer patients in The Cancer Genome Atlas (TCGA). Statistically significant gene pairs associated with survival were identified from multiple genomic profiles, which include many individual genes that have weak or no effect on survival. Moreover, we also show that integrated networks, constructed by merging networks from multiple genomic profiles, demonstrate better topological properties and biological significance than individual networks. CONCLUSIONS We have developed a simple but powerful analysis tool that is able to detect gene-gene interactions associated with clinical outcome on multiple genomic profiles. By being network-based, our approach provides a better insight into the underlying gene-gene interaction mechanisms that affect the clinical outcome of cancer patients.
Collapse
Affiliation(s)
- Hyun-Hwan Jeong
- Department of Information and Computer Engineering, Ajou University, Suwon, 443-749, Republic of Korea.
| | - Sangseob Leem
- Department of Information and Computer Engineering, Ajou University, Suwon, 443-749, Republic of Korea.
| | - Kyubum Wee
- Department of Information and Computer Engineering, Ajou University, Suwon, 443-749, Republic of Korea.
| | - Kyung-Ah Sohn
- Department of Information and Computer Engineering, Ajou University, Suwon, 443-749, Republic of Korea.
| |
Collapse
|
34
|
Brachova P, Mueting SR, Carlson MJ, Goodheart MJ, Button AM, Mott SL, Dai D, Thiel KW, Devor EJ, Leslie KK. TP53 oncomorphic mutations predict resistance to platinum‑ and taxane‑based standard chemotherapy in patients diagnosed with advanced serous ovarian carcinoma. Int J Oncol 2014; 46:607-18. [PMID: 25385265 PMCID: PMC4277253 DOI: 10.3892/ijo.2014.2747] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/20/2014] [Indexed: 12/22/2022] Open
Abstract
Individual mutations in the tumor suppressor TP53 alter p53 protein function. Some mutations create a non-functional protein, whereas others confer oncogenic activity, which we term ‘oncomorphic’. Since mutations in TP53 occur in nearly all ovarian tumors, the objective of this study was to determine the relationship of oncomorphic TP53 mutations with patient outcomes in advanced serous ovarian cancer patients. Clinical and molecular data from 264 high-grade serous ovarian cancer patients uniformly treated with standard platinum- and taxane-based adjuvant chemotherapy were downloaded from The Cancer Genome Atlas (TCGA) portal. Additionally, patient samples were obtained from the University of Iowa and individual mutations were analyzed in ovarian cancer cell lines. Mutations in the TP53 were annotated and categorized as oncomorphic, loss of function (LOF), or unclassified. Associations between mutation types, chemoresistance, recurrence, and progression-free survival (PFS) were calculated. Oncomorphic TP53 mutations were present in 21.3% of ovarian cancers in the TCGA dataset. Patients with oncomorphic TP53 mutations demonstrated significantly worse PFS, a 60% higher risk of recurrence (HR=1.60, 95% confidence intervals 1.09, 2.33, p=0.015), and higher rates of platinum resistance (χ2 test p=0.0024) when compared with single nucleotide mutations not categorized as oncomorphic. Furthermore, tumors containing oncomorphic TP53 mutations displayed unique protein expression profiles, and some mutations conferred increased clonogenic capacity in ovarian cancer cell models. Our study reveals that oncomorphic TP53 mutations are associated with worse patient outcome. These data suggest that future studies should take into consideration the functional consequences of TP53 mutations when determining treatment options.
Collapse
Affiliation(s)
- Pavla Brachova
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA
| | - Samuel R Mueting
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA
| | - Matthew J Carlson
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA
| | - Michael J Goodheart
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA
| | - Anna M Button
- Holden Comprehensive Cancer Center and Molecular and Cellular Biology Program, University of Iowa, Iowa City, IA 52242, USA
| | - Sarah L Mott
- Holden Comprehensive Cancer Center and Molecular and Cellular Biology Program, University of Iowa, Iowa City, IA 52242, USA
| | - Donghai Dai
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA
| | - Kristina W Thiel
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA
| | - Eric J Devor
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA
| | - Kimberly K Leslie
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
35
|
Ezzati M, Abdullah A, Shariftabrizi A, Hou J, Kopf M, Stedman JK, Samuelson R, Shahabi S. Recent Advancements in Prognostic Factors of Epithelial Ovarian Carcinoma. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:953509. [PMID: 27382614 PMCID: PMC4897239 DOI: 10.1155/2014/953509] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/26/2014] [Indexed: 01/03/2023]
Abstract
Ovarian cancer remains the most common cause of gynecologic cancer-related death among women in developed countries. Nevertheless, subgroups of ovarian cancer patients experience relatively longer survival. Efforts to identify prognostic factors that characterize such patients are ongoing, with investigational areas including tumor characteristics, surgical management, inheritance patterns, immunologic factors, and genomic patterns. This review discusses various demographic, clinical, and molecular factors implicating longevity and ovarian cancer survival. Continued efforts at identifying these prognosticators may result in invaluable adjuncts to the treatment of ovarian cancer, with the ultimate goal of advancing patient care.
Collapse
Affiliation(s)
- Mohammad Ezzati
- Department of Obstetrics and Gynecology, Washington Hospital Center, 110 Irving Street NW, Washington, DC 20010, USA
- Department of Obstetrics and Gynecology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Amer Abdullah
- Department of Obstetrics, Gynecology and Reproductive Biology, Danbury Hospital, 24 Hospital Avenue, Danbury, CT 06810, USA
| | - Ahmad Shariftabrizi
- Department of Pathology and Laboratory Medicine, School of Medicine, Tufts University, 800 Washington Street, Boston, MA 02111, USA
| | - June Hou
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine and Montefiore Medical Center, Montefiore Centennial, 3332 Rochambeau Avenue, Bronx, NY 10467-2836, USA
| | - Michael Kopf
- Department of Medicine, Danbury Hospital, 24 Hospital Avenue, Danbury, CT 06810, USA
| | - Jennifer K. Stedman
- Department of Obstetrics, Gynecology and Reproductive Biology, Danbury Hospital, 24 Hospital Avenue, Danbury, CT 06810, USA
| | - Robert Samuelson
- Department of Obstetrics, Gynecology and Reproductive Biology, Danbury Hospital, 24 Hospital Avenue, Danbury, CT 06810, USA
| | - Shohreh Shahabi
- Department of Obstetrics, Gynecology and Reproductive Biology, Danbury Hospital, 24 Hospital Avenue, Danbury, CT 06810, USA
| |
Collapse
|
36
|
p53 protein aggregation promotes platinum resistance in ovarian cancer. Oncogene 2014; 34:3605-16. [PMID: 25263447 DOI: 10.1038/onc.2014.296] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/25/2014] [Accepted: 07/31/2014] [Indexed: 12/24/2022]
Abstract
High-grade serous ovarian carcinoma (HGSOC), the most lethal gynecological cancer, often leads to chemoresistant diseases. The p53 protein is a key transcriptional factor regulating cellular homeostasis. A majority of HGSOCs have inactive p53 because of genetic mutations. However, genetic mutation is not the only cause of p53 inactivation. The aggregation of p53 protein has been discovered in different types of cancers and may be responsible for impairing the normal transcriptional activation and pro-apoptotic functions of p53. We demonstrated that in a unique population of HGSOC cancer cells with cancer stem cell properties, p53 protein aggregation is associated with p53 inactivation and platinum resistance. When these cancer stem cells differentiated into their chemosensitive progeny, they lost tumor-initiating capacity and p53 aggregates. In addition to the association of p53 aggregation and chemoresistance in HGSOC cells, we further demonstrated that the overexpression of a p53-positive regulator, p14ARF, inhibited MDM2-mediated p53 degradation and led to the imbalance of p53 turnover that promoted the formation of p53 aggregates. With in vitro and in vivo models, we demonstrated that the inhibition of p14ARF could suppress p53 aggregation and sensitize cancer cells to platinum treatment. Moreover, by two-dimensional gel electrophoresis and mass spectrometry we discovered that the aggregated p53 may function uniquely by interacting with proteins that are critical for cancer cell survival and tumor progression. Our findings help us understand the poor chemoresponse of a subset of HGSOC patients and suggest p53 aggregation as a new marker for chemoresistance. Our findings also suggest that inhibiting p53 aggregation can reactivate p53 pro-apoptotic function. Therefore, p53 aggregation is a potential therapeutic target for reversing chemoresistance. This is paramount for improving ovarian cancer patients' responses to chemotherapy, and thus increasing their survival rate.
Collapse
|
37
|
Masica DL, Li S, Douville C, Manola J, Ferris RL, Burtness B, Forastiere AA, Koch WM, Chung CH, Karchin R. Predicting survival in head and neck squamous cell carcinoma from TP53 mutation. Hum Genet 2014; 134:497-507. [PMID: 25108461 DOI: 10.1007/s00439-014-1470-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 07/17/2014] [Indexed: 12/20/2022]
Abstract
For TP53-mutated head and neck squamous cell carcinomas (HNSCCs), the codon and specific amino acid sequence change resulting from a patient's mutation can be prognostic. Thus, developing a framework to predict patient survival for specific mutations in TP53 would be valuable. There are many bioinformatics and functional methods for predicting the phenotypic impact of genetic variation, but their overall clinical value remains unclear. Here, we assess the ability of 15 different methods to predict HNSCC patient survival from TP53 mutation, using TP53 mutation and clinical data from patients enrolled in E4393 by the Eastern Cooperative Oncology Group (ECOG), which investigated whether TP53 mutations in surgical margins were predictive of disease recurrence. These methods include: server-based computational tools SIFT, PolyPhen-2, and Align-GVGD; our in-house POSE and VEST algorithms; the rules devised in Poeta et al. with and without considerations for splice-site mutations; location of mutation in the DNA-bound TP53 protein structure; and a functional assay measuring WAF1 transactivation in TP53-mutated yeast. We assessed method performance using overall survival (OS) and progression-free survival (PFS) from 420 HNSCC patients, of whom 224 had TP53 mutations. Each mutation was categorized as "disruptive" or "non-disruptive". For each method, we compared the outcome between the disruptive group vs. the non-disruptive group. The rules devised by Poeta et al. with or without our splice-site modification were observed to be superior to others. While the differences in OS (disruptive vs. non-disruptive) appear to be marginally significant (Poeta rules + splice rules, P = 0.089; Poeta rules, P = 0.053), both algorithms identified the disruptive group as having significantly worse PFS outcome (Poeta rules + splice rules, P = 0.011; Poeta rules, P = 0.027). In general, prognostic performance was low among assessed methods. Further studies are required to develop and validate methods that can predict functional and clinical significance of TP53 mutations in HNSCC patients.
Collapse
Affiliation(s)
- David L Masica
- Department of Biomedical Engineering, Institute for Computational Medicine, The Johns Hopkins University, Baltimore, MD, USA,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Mir R, Stanzani E, Martinez-Soler F, Villanueva A, Vidal A, Condom E, Ponce J, Gil J, Tortosa A, Giménez-Bonafé P. YM155 sensitizes ovarian cancer cells to cisplatin inducing apoptosis and tumor regression. Gynecol Oncol 2013; 132:211-20. [PMID: 24262875 DOI: 10.1016/j.ygyno.2013.11.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/05/2013] [Accepted: 11/11/2013] [Indexed: 11/29/2022]
Abstract
OBJECTIVE The objective of this study is to chemosensitize ovarian cancer (OVCa) cells to cisplatin (CDDP) using an inhibitor of Survivin, YM155. The efficacy of YM155 in combination with CDDP was determined in vitro, ex vivo and in vivo. METHODS Human OVCa cell lines A2780p and their cisplatin-resistant derivative A2780cis, were treated with CDDP, YM155, and the combined treatment (YM155+CDDP), and cell viability, mRNA and protein expression levels, cell-cycle distribution, and DNA damage were then evaluated. Furthermore, the efficacy of YM155 combined with CDDP was further examined in established primary cell cultures and xenograft models. RESULTS The combination of YM155 with CDDP induced G2/M cell cycle arrest and apoptosis, increased DNA damage, and decreased Survivin levels, especially in A2780cis CDDP-resistant cells. Additionally, YM155 in combination with CDDP sensitized primary cell cultures to CDDP. Studies in vivo showed how this combination significantly decreased the tumor size of OVCa xenografts. CONCLUSIONS Our results demonstrate that in OVCa cells the expression of Survivin did not affect their sensitivity to YM155, suggesting that Survivin was not the only target of YM155. The combination of YM155 with CDDP could be a good option for therapy of CDDP-resistant OVCa, independently of p53 status.
Collapse
Affiliation(s)
- Roser Mir
- Departament de Ciències Fisiològiques II, Faculty of Medicine, Campus of Health Sciences of Bellvitge, Universitat de Barcelona, IDIBELL, Spain
| | - Elisabetta Stanzani
- Departament de Ciències Fisiològiques II, Faculty of Medicine, Campus of Health Sciences of Bellvitge, Universitat de Barcelona, IDIBELL, Spain
| | - Fina Martinez-Soler
- Departament de Ciències Fisiològiques II, Faculty of Medicine, Campus of Health Sciences of Bellvitge, Universitat de Barcelona, IDIBELL, Spain; Department of Basic Nursing, School of Nursing of the Health Campus of Bellvitge, Universitat de Barcelona, IDIBELL, Spain
| | - Alberto Villanueva
- Translational Research Laboratory, Catalan Institute of Oncology, Bellvitge Biomedical Research Institute, IDIBELL, Spain
| | - August Vidal
- Department of Pathology, Hospital de Bellvitge, Department of Pathology and Experimental Therapeutics, Universitat de Barcelona, IDIBELL, Spain
| | - Enric Condom
- Department of Pathology, Hospital de Bellvitge, Department of Pathology and Experimental Therapeutics, Universitat de Barcelona, IDIBELL, Spain
| | - Jordi Ponce
- Department of Gynecology, Hospital de Bellvitge, Barcelona, Spain
| | - Joan Gil
- Departament de Ciències Fisiològiques II, Faculty of Medicine, Campus of Health Sciences of Bellvitge, Universitat de Barcelona, IDIBELL, Spain
| | - Avelina Tortosa
- Department of Basic Nursing, School of Nursing of the Health Campus of Bellvitge, Universitat de Barcelona, IDIBELL, Spain.
| | - Pepita Giménez-Bonafé
- Departament de Ciències Fisiològiques II, Faculty of Medicine, Campus of Health Sciences of Bellvitge, Universitat de Barcelona, IDIBELL, Spain.
| |
Collapse
|