1
|
Machan S, Rodríguez M, Manso R, Borregón J, Chamizo C, Alonso-Alonso R, Rodríguez-Peralto JL, Torres Nieto MÁ, Monteagudo C, García Toro E, Cerroni L, García C, Estrach T, García Herrera A, Ferrer B, García-Patos V, Segues N, Díaz de la Pinta FJ, Afonso-Martin JL, Peñate Y, Limeres-Gonzalez MÁ, González-Núñez MÁ, González-Cruz C, García Fernández E, Cereceda L, Minguez P, de la Fuente L, Requena L, Rodríguez-Pinilla SM. Different Mutational Profiles of Subcutaneous Panniculitis-like T-cell Lymphoma and Lupus Panniculitis: An Additional Case Series. ACTAS DERMO-SIFILIOGRAFICAS 2024:S0001-7310(24)00595-7. [PMID: 39032781 DOI: 10.1016/j.ad.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 05/28/2024] [Accepted: 06/29/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Subcutaneous panniculitis-like T-cell lymphoma (SPTCL) is a rare cytotoxic T-cell lymphoma with indolent behavior, mostly present in women and associated with immunological diseases whose pathogenic background is still poorly understood. SPTCL is associated with lupus erythematosus panniculitis (LEP) and histologically misdiagnosed. OBJECTIVES The aim of our study was to identify mutations affecting the pathogenesis of both SPTCL and LEP. MATERIALS AND METHODS We studied a total of 10 SPTCL and 10 LEP patients using targeted next-generation sequencing and pyrosequencing. Differences in gene expression between molecular subgroups were investigated using NanoString technology. Clinical data were collected, and correlations sought with the molecular data obtained. RESULTS The mutational profile of SPTCL and LEP is different. We identified fewer pathogenic mutations than previously reported in SPTCL, noting a single HAVCR2-mutated SPTCL case. Interestingly, 40% of our SPTCL cases showed the pathogenic TP53 (p.Pro72Arg) (P72R) variant. Although cases showing HAVCR2 mutations or the TP53 (P72R) variant had more severe symptomatic disease, none developed hemophagocytic syndrome (HPS). Furthermore, TP53 (P72R)-positive cases were characterized by a lower metabolic signaling pathway and higher levels of CD28 expression and Treg signaling genes. In addition, 30% of our cases featured the same mutation (T735C) of the epigenetic modificatory gene DNMT3A. None of the LEP cases showed mutations in any of the studied genes. CONCLUSIONS The mutational landscape of SPTCL is broader than previously anticipated. We describe, for the first time, the involvement of the TP53 (P72R) pathogenic variant in this subgroup of tumors, consider the possible role of different genetic backgrounds in the development of SPTCL, and conclude that LEP does not follow the same pathogenic pathway as SPTCL.
Collapse
Affiliation(s)
- S Machan
- Department of Dermatology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain
| | - M Rodríguez
- Department of Pathology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain
| | - R Manso
- Department of Pathology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain.
| | - J Borregón
- Department of Pathology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain
| | - C Chamizo
- Department of Pathology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain
| | - R Alonso-Alonso
- Department of Pathology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain
| | | | | | - C Monteagudo
- Hospital Clínico Universitario de Valencia, Universidad de Valencia, Valencia, Spain
| | | | - L Cerroni
- Dermatopathology Research Unit, Department of Dermatology, Medical University of Graz, Graz, Austria
| | - C García
- Hospital Universitario de Canarias, Tenerife, Spain
| | - T Estrach
- Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - B Ferrer
- Hospital Vall d'Hebron, Barcelona, Spain
| | | | - N Segues
- Hospital Universitario Donostia, San Sebastián, Spain
| | - F J Díaz de la Pinta
- Department of Pathology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain
| | - J L Afonso-Martin
- Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain
| | - Y Peñate
- Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain
| | - M Á Limeres-Gonzalez
- Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria, Spain
| | - M Á González-Núñez
- Hospital Ciudad de Coria y Hospital San Pedro de Alcántara, Cáceres, Spain
| | | | - E García Fernández
- Department of Hematology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain
| | - L Cereceda
- Department of Pathology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain
| | - P Minguez
- Department of Genetics, Instituto de Investigación Sanitaria - Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain; Bioinformatics Unit, Instituto de Investigación Sanitaria - Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - L de la Fuente
- Department of Genetics, Instituto de Investigación Sanitaria - Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain; Bioinformatics Unit, Instituto de Investigación Sanitaria - Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - L Requena
- Department of Dermatology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain
| | - S M Rodríguez-Pinilla
- Department of Pathology, Fundación Jiménez Díaz-IIS, Universidad Autónoma de Madrid, Madrid, Spain; CIBERONC (Centro de Investigación Biomédica en Red de Cáncer), Spain
| |
Collapse
|
2
|
Wenzl K, Stokes ME, Novak JP, Bock AM, Khan S, Hopper MA, Krull JE, Dropik AR, Walker JS, Sarangi V, Mwangi R, Ortiz M, Stong N, Huang CC, Maurer MJ, Rimsza L, Link BK, Slager SL, Asmann Y, Mondello P, Morin R, Ansell SM, Habermann TM, Witzig TE, Feldman AL, King RL, Nowakowski G, Cerhan JR, Gandhi AK, Novak AJ. Multiomic analysis identifies a high-risk signature that predicts early clinical failure in DLBCL. Blood Cancer J 2024; 14:100. [PMID: 38902256 PMCID: PMC11189905 DOI: 10.1038/s41408-024-01080-0] [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: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/22/2024] Open
Abstract
Recent genetic and molecular classification of DLBCL has advanced our knowledge of disease biology, yet were not designed to predict early events and guide anticipatory selection of novel therapies. To address this unmet need, we used an integrative multiomic approach to identify a signature at diagnosis that will identify DLBCL at high risk of early clinical failure. Tumor biopsies from 444 newly diagnosed DLBCL were analyzed by WES and RNAseq. A combination of weighted gene correlation network analysis and differential gene expression analysis was used to identify a signature associated with high risk of early clinical failure independent of IPI and COO. Further analysis revealed the signature was associated with metabolic reprogramming and identified cases with a depleted immune microenvironment. Finally, WES data was integrated into the signature and we found that inclusion of ARID1A mutations resulted in identification of 45% of cases with an early clinical failure which was validated in external DLBCL cohorts. This novel and integrative approach is the first to identify a signature at diagnosis, in a real-world cohort of DLBCL, that identifies patients at high risk for early clinical failure and may have significant implications for design of therapeutic options.
Collapse
Affiliation(s)
- Kerstin Wenzl
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Matthew E Stokes
- Informatics and Predictive Sciences, , Bristol Myers Squibb, Summit, NJ, USA
| | | | | | - Sana Khan
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Vivekananda Sarangi
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Raphael Mwangi
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Maria Ortiz
- Informatics and Predictive Sciences, Celgene Institute for Translational Research Europe (CITRE), Seville, Spain
| | - Nicholas Stong
- Informatics and Predictive Sciences, , Bristol Myers Squibb, Summit, NJ, USA
| | - C Chris Huang
- Translational Medicine Hematology, Bristol Myers Squibb, Summit, NJ, USA
| | - Matthew J Maurer
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Lisa Rimsza
- Division of Hematopathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Brian K Link
- Division of Hematology, University of Iowa, Iowa, USA
| | - Susan L Slager
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Yan Asmann
- Department of Quantitative Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | | - Ryan Morin
- Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | | | | | | | - Andrew L Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Rebecca L King
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - James R Cerhan
- Department of Quantitative Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Anita K Gandhi
- Translational Medicine Hematology, Bristol Myers Squibb, Summit, NJ, USA
| | - Anne J Novak
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Ganci F, Allegretti M, Frascolla C, Spinella F, Rollo F, Sacconi A, Valentina PD, Palcau AC, Manciocco V, Vescovo M, Cotroneo E, Blandino F, Benevolo M, Covello R, Muti P, Strano S, Vidiri A, Fontemaggi G, Pellini R, Blandino G. Combined TP53 status in tumor-free resection margins and circulating microRNA profiling predicts the risk of locoregional recurrence in head and neck cancer. Biomark Res 2024; 12:32. [PMID: 38444004 PMCID: PMC10916059 DOI: 10.1186/s40364-024-00576-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/15/2024] [Indexed: 03/07/2024] Open
Abstract
Locoregional recurrences represent a frequently unexpected problem in head and neck squamous cell carcinoma (HNSCC). Relapse often (10-30%) occurs in patients with histologically negative resection margins (RMs), probably due to residual tumor cells or hidden pre-cancerous lesions in normal mucosa, both missed by histopathological examination. Therefore, definition of a 'clean' or tumor-negative RM is controversial, demanding for novel approaches to be accurately explored. Here, we evaluated next generation sequencing (NGS) and digital PCR (dPCR) as tools to profile TP53 mutational status and circulating microRNA expression aiming at scoring the locoregional risk of recurrence by means of molecular analyses. Serial monitoring of these biomarkers allowed identifying patients at high risk, laying the ground for accurate tracking of disease evolution and potential intensification of post-operative treatments. Additionally, our pipeline demonstrated its applicability into the clinical routine, being cost-effective and feasible in terms of patient sampling, holding promise to accurately (re)-stage RMs in the era of precision medicine.
Collapse
Affiliation(s)
- Federica Ganci
- Translational Oncologic Research Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Matteo Allegretti
- Translational Oncologic Research Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Carlotta Frascolla
- Translational Oncologic Research Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Francesca Spinella
- Department of Research and Development, Eurofins Genoma Group, Rome, Italy
| | - Francesca Rollo
- Pathology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Andrea Sacconi
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Pascale De Valentina
- Translational Oncologic Research Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Alina Catalina Palcau
- Translational Oncologic Research Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Valentina Manciocco
- Otolaryngology-Head and Neck Surgery, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Mariavittoria Vescovo
- Pathology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Ettore Cotroneo
- Clinical and Technical Department Management, Eurofins Genoma Group, Rome, Italy
| | - Francesca Blandino
- Department of Research and Development, Eurofins Genoma Group, Rome, Italy
| | - Maria Benevolo
- Pathology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Renato Covello
- Pathology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Paola Muti
- Department of Health Research Methods, Evidence, and Impact, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Sabrina Strano
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Giulia Fontemaggi
- Translational Oncologic Research Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Raul Pellini
- Otolaryngology-Head and Neck Surgery, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Giovanni Blandino
- Translational Oncologic Research Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| |
Collapse
|
4
|
Zhang X, Bolck HA, Rupp NJ, Moch H. Genomic alterations and diagnosis of renal cancer. Virchows Arch 2024; 484:323-337. [PMID: 37999735 PMCID: PMC10948545 DOI: 10.1007/s00428-023-03700-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/24/2023] [Accepted: 11/04/2023] [Indexed: 11/25/2023]
Abstract
The application of molecular profiling has made substantial impact on the classification of urogenital tumors. Therefore, the 2022 World Health Organization incorporated the concept of molecularly defined renal tumor entities into its classification, including succinate dehydrogenase-deficient renal cell carcinoma (RCC), FH-deficient RCC, TFE3-rearranged RCC, TFEB-altered RCC, ALK-rearranged RCC, ELOC-mutated RCC, and renal medullary RCC, which are characterized by SMARCB1-deficiency. This review aims to provide an overview of the most important molecular alterations in renal cancer, with a specific focus on the diagnostic value of characteristic genomic aberrations, their chromosomal localization, and associations with renal tumor subtypes. It may not yet be the time to completely shift to a molecular RCC classification, but undoubtedly, the application of molecular profiling will enhance the accuracy of renal cancer diagnosis, and ultimately guide personalized treatment strategies for patients.
Collapse
Affiliation(s)
- Xingming Zhang
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstr. 12, 8091, Zurich, Switzerland
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Hella A Bolck
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstr. 12, 8091, Zurich, Switzerland
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstr. 12, 8091, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstr. 12, 8091, Zurich, Switzerland.
- Faculty of Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Gu X, Huang Z, Chen J, Luo Y, Ge S, Jia R, Song X, Chai P, Xu S, Fan X. Establishment and Characterization of a TP53-Mutated Eyelid Sebaceous Carcinoma Cell Line. Invest Ophthalmol Vis Sci 2023; 64:16. [PMID: 38095907 PMCID: PMC10723222 DOI: 10.1167/iovs.64.15.16] [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: 09/14/2023] [Accepted: 11/19/2023] [Indexed: 12/17/2023] Open
Abstract
Purpose Eyelid sebaceous carcinoma (SeC) is the third most frequent eyelid malignancy worldwide and is relatively prevalent in Asian patients. An eyelid SeC cell line model is necessary for experimental research to explore the etiology and pathogenesis of eyelid SeC. This study established and characterized an eyelid SeC cell line with a TP53 mutation that might be useful for analyzing potential treatment options for eyelid SeC. Methods The eyelid SeC cell line SHNPH-SeC was obtained from a patient with eyelid SeC at Shanghai Ninth People's Hospital (SHNPH), Shanghai JiaoTong University School of Medicine. Immunofluorescence staining was employed to detect the origination and proliferation activity. Short tandem repeat (STR) profiling was performed for verification. Chromosome analysis was implemented to investigate chromosome aberrations. Whole exome sequencing (WES) was used to discover genomic mutations. Cell proliferation assays were performed to identify sensitivity to mitomycin-C (MMC) and 5-fluorouracil (5-FU). Results SHNPH-SeC cells were successively subcultured for more than 100 passages and demonstrated rapid proliferation and migration. Karyotype analysis revealed abundant chromosome aberrations, and WES revealed SeC-related mutations in TP53, KMT2C, and ERBB2. An in vivo tumor model was successfully established in NOD/SCID mice. Biomarkers of eyelid SeC, including cytokeratin 5 (CK5), epithelial membrane antigen (EMA), adipophilin, p53, and Ki-67, were detected in SHNPH-SeC cells, original tumors, and xenografts. MMC and 5-FU inhibited the proliferation and migration of SHNPH-SeC cells, and SHNPH-SeC cells presented a greater drug response than non-TP53-mutated SeC cells. Conclusions The newly established eyelid SeC cell line SHNPH-SeC demonstrates mutation in TP53, the most commonly mutated gene in SeC. It presents SeC properties and malignant characteristics that may facilitate the investigation of cellular behaviors and molecular mechanisms of SeC to explore promising therapeutic strategies.
Collapse
Affiliation(s)
- Xiang Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Ziyue Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jie Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yingxiu Luo
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xin Song
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Shiqiong Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
6
|
Deng Z, Shen D, Yu M, Zhou F, Shan D, Fang Y, Jin W, Qian K, Li S, Wang G, Zhang Y, Ju L, Xiao Y, Wang X. Pectolinarigenin inhibits bladder urothelial carcinoma cell proliferation by regulating DNA damage/autophagy pathways. Cell Death Discov 2023; 9:214. [PMID: 37393350 DOI: 10.1038/s41420-023-01508-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/25/2023] [Accepted: 06/20/2023] [Indexed: 07/03/2023] Open
Abstract
Pectolinarigenin (PEC), an active compound isolated from traditional herbal medicine, has shown potential anti-tumor properties against various types of cancer cells. However, its mechanism of action in bladder cancer (BLCA), which is one of the fatal human carcinomas, remains unexplored. In this study, we first revealed that PEC, as a potential DNA topoisomerase II alpha (TOP2A) poison, can target TOP2A and cause significant DNA damage. PEC induced G2/M phase cell cycle arrest via p53 pathway. Simultaneously, PEC can perform its unique function by inhibiting the late autophagic flux. The blocking of autophagy caused proliferation inhibition of BLCA and further enhanced the DNA damage effect of PEC. In addition, we proved that PEC could intensify the cytotoxic effect of gemcitabine (GEM) on BLCA cells in vivo and in vitro. Summarily, we first systematically revealed that PEC had great potential as a novel TOP2A poison and an inhibitor of late autophagic flux in treating BLCA.
Collapse
Affiliation(s)
- Zhao Deng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dexin Shen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mengxue Yu
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
| | - Fenfang Zhou
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Danni Shan
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
| | - Yayun Fang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
| | - Wan Jin
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Euler Technology, ZGC Life Sciences Park, Beijing, China
| | - Kaiyu Qian
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shenjuan Li
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Medical Research Institute, Wuhan University, Wuhan, China
| | - Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
| | - Yi Zhang
- Euler Technology, ZGC Life Sciences Park, Beijing, China
- Center for Quantitative Biology, School of Life Sciences, Peking University, Beijing, China
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Yu Xiao
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Urological Diseases, Wuhan University, Wuhan, China.
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Medical Research Institute, Wuhan University, Wuhan, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
7
|
Wenzl K, Stokes M, Novak JP, Bock AM, Khan S, Hopper MA, Krull JE, Dropik AR, Walker JS, Sarangi V, Mwangi R, Ortiz M, Stong N, Huang CC, Maurer MJ, Rimsza L, Link BK, Slager SL, Asmann Y, Mondello P, Morin R, Ansell SM, Habermann TM, Feldman AL, King RL, Nowakowski G, Cerhan JR, Gandhi AK, Novak AJ. Multiomic Analysis Identifies a High-Risk Metabolic and TME Depleted Signature that Predicts Early Clinical Failure in DLBCL. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.07.23290748. [PMID: 37333387 PMCID: PMC10274962 DOI: 10.1101/2023.06.07.23290748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
PURPOSE 60-70% of newly diagnosed diffuse large B-cell lymphoma (DLBCL) patients avoid events within 24 months of diagnosis (EFS24) and the remainder have poor outcomes. Recent genetic and molecular classification of DLBCL has advanced our knowledge of disease biology, yet were not designed to predict early events and guide anticipatory selection of novel therapies. To address this unmet need, we used an integrative multiomic approach to identify a signature at diagnosis that will identify DLBCL at high risk of early clinical failure. PATIENTS AND METHODS Tumor biopsies from 444 newly diagnosed DLBCL were analyzed by WES and RNAseq. A combination of weighted gene correlation network analysis and differential gene expression analysis followed by integration with clinical and genomic data was used to identify a multiomic signature associated with high risk of early clinical failure. RESULTS Current DLBCL classifiers are unable to discriminate cases who fail EFS24. We identified a high risk RNA signature that had a hazard ratio (HR, 18.46 [95% CI 6.51-52.31] P < .001) in a univariate model, which did not attenuate after adjustment for age, IPI and COO (HR, 20.8 [95% CI, 7.14-61.09] P < .001). Further analysis revealed the signature was associated with metabolic reprogramming and a depleted immune microenvironment. Finally, WES data was integrated into the signature and we found that inclusion of ARID1A mutations resulted in identification of 45% of cases with an early clinical failure which was validated in external DLBCL cohorts. CONCLUSION This novel and integrative approach is the first to identify a signature at diagnosis that will identify DLBCL at high risk for early clinical failure and may have significant implications for design of therapeutic options.
Collapse
|
8
|
Guo Y, Tang Y, Lu G, Gu J. p53 at the Crossroads between Doxorubicin-Induced Cardiotoxicity and Resistance: A Nutritional Balancing Act. Nutrients 2023; 15:nu15102259. [PMID: 37242146 DOI: 10.3390/nu15102259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/19/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Doxorubicin (DOX) is a highly effective chemotherapeutic drug, but its long-term use can cause cardiotoxicity and drug resistance. Accumulating evidence demonstrates that p53 is directly involved in DOX toxicity and resistance. One of the primary causes for DOX resistance is the mutation or inactivation of p53. Moreover, because the non-specific activation of p53 caused by DOX can kill non-cancerous cells, p53 is a popular target for reducing toxicity. However, the reduction in DOX-induced cardiotoxicity (DIC) via p53 suppression is often at odds with the antitumor advantages of p53 reactivation. Therefore, in order to increase the effectiveness of DOX, there is an urgent need to explore p53-targeted anticancer strategies owing to the complex regulatory network and polymorphisms of the p53 gene. In this review, we summarize the role and potential mechanisms of p53 in DIC and resistance. Furthermore, we focus on the advances and challenges in applying dietary nutrients, natural products, and other pharmacological strategies to overcome DOX-induced chemoresistance and cardiotoxicity. Lastly, we present potential therapeutic strategies to address key issues in order to provide new ideas for increasing the clinical use of DOX and improving its anticancer benefits.
Collapse
Affiliation(s)
- Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
9
|
Adams CM, Mitra R, Xiao Y, Michener P, Palazzo J, Chao A, Gour J, Cassel J, Salvino JM, Eischen CM. Targeted MDM2 Degradation Reveals a New Vulnerability for p53-Inactivated Triple-Negative Breast Cancer. Cancer Discov 2023; 13:1210-1229. [PMID: 36734633 PMCID: PMC10164114 DOI: 10.1158/2159-8290.cd-22-1131] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/29/2022] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
Triple-negative breast cancers (TNBC) frequently inactivate p53, increasing their aggressiveness and therapy resistance. We identified an unexpected protein vulnerability in p53-inactivated TNBC and designed a new PROteolysis TArgeting Chimera (PROTAC) to target it. Our PROTAC selectively targets MDM2 for proteasome-mediated degradation with high-affinity binding and VHL recruitment. MDM2 loss in p53 mutant/deleted TNBC cells in two-dimensional/three-dimensional culture and TNBC patient explants, including relapsed tumors, causes apoptosis while sparing normal cells. Our MDM2-PROTAC is stable in vivo, and treatment of TNBC xenograft-bearing mice demonstrates tumor on-target efficacy with no toxicity to normal cells, significantly extending survival. Transcriptomic analyses revealed upregulation of p53 family target genes. Investigations showed activation and a required role for TAp73 to mediate MDM2-PROTAC-induced apoptosis. Our data, challenging the current MDM2/p53 paradigm, show MDM2 is required for p53-inactivated TNBC cell survival, and PROTAC-targeted MDM2 degradation is an innovative potential therapeutic strategy for TNBC and superior to existing MDM2 inhibitors. SIGNIFICANCE p53-inactivated TNBC is an aggressive, therapy-resistant, and lethal breast cancer subtype. We designed a new compound targeting an unexpected vulnerability we identified in TNBC. Our MDM2-targeted degrader kills p53-inactivated TNBC cells, highlighting the requirement for MDM2 in TNBC cell survival and as a new therapeutic target for this disease. See related commentary by Peuget and Selivanova, p. 1043. This article is highlighted in the In This Issue feature, p. 1027.
Collapse
Affiliation(s)
- Clare M. Adams
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ramkrishna Mitra
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Peter Michener
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Juan Palazzo
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Allen Chao
- The Wistar Institute, Philadelphia, PA, USA
| | | | | | | | - Christine M. Eischen
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
10
|
Zhang M, Xiao F, Li Y, Chen Z, Zhang X, Zhang X, Song J, Zhang Y, Si X, Bai J, Yagüe E, Zhou Y. The miR-106b-25 cluster mediates drug resistance in myeloid leukaemias by inactivating multiple apoptotic genes. Int J Hematol 2023; 117:236-250. [PMID: 36399285 DOI: 10.1007/s12185-022-03483-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/19/2022]
Abstract
Drug resistance is a major obstacle to the successful treatment of cancer. The role of the miR-106b-25 cluster in drug resistance of haematologic malignancies has not yet been elucidated. Here, we show that the miR-106b-25 cluster mediates resistance to therapeutic agents with structural and mechanistic dissimilarity in vitro and in vivo. RNA sequencing data revealed that overexpression of the miR-106b-25 cluster or its individual miRNAs resulted in downregulation of multiple key regulators of apoptotic pathways. Luciferase reporter assay identified TP73 as a direct target of miR-93 and miR-106b, BAK1 as a direct target of miR-25 and CASP7 as a direct target of all three miRNAs. We also showed that inhibitors of the miR-106b-25 cluster and BCL-2 exert synergistic effects on apoptosis induction in primary myeloid leukaemic cells. Thus, the members of the miR-106b-25 cluster may jointly contribute to myeloid leukaemia drug resistance by inactivating multiple apoptotic genes. Targeting this cluster could be a promising combination strategy in patients resistant to therapeutic agents that induce apoptosis.
Collapse
Affiliation(s)
- Mingying Zhang
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Fangnan Xiao
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Yunan Li
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Zizhen Chen
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Xiaoyun Zhang
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Xiaoru Zhang
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Junzhe Song
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Yuhui Zhang
- Department of Hematology, The Second Affiliated Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Xiaohui Si
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jie Bai
- Department of Hematology, The Second Affiliated Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Ernesto Yagüe
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0NN, UK.
| | - Yuan Zhou
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China.
| |
Collapse
|
11
|
Deciphering the Role of p53 and TAp73 in Neuroblastoma: From Pathogenesis to Treatment. Cancers (Basel) 2022; 14:cancers14246212. [PMID: 36551697 PMCID: PMC9777536 DOI: 10.3390/cancers14246212] [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: 11/04/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Neuroblastoma (NB) is an embryonic cancer that develops from neural crest stem cells, being one of the most common malignancies in children. The clinical manifestation of this disease is highly variable, ranging from spontaneous regression to increased aggressiveness, which makes it a major therapeutic challenge in pediatric oncology. The p53 family proteins p53 and TAp73 play a key role in protecting cells against genomic instability and malignant transformation. However, in NB, their activities are commonly inhibited by interacting proteins such as murine double minute (MDM)2 and MDMX, mutant p53, ΔNp73, Itch, and Aurora kinase A. The interplay between the p53/TAp73 pathway and N-MYC, a known biomarker of poor prognosis and drug resistance in NB, also proves to be decisive in the pathogenesis of this tumor. More recently, a strong crosstalk between microRNAs (miRNAs) and p53/TAp73 has been established, which has been the focused of great attention because of its potential for developing new therapeutic strategies. Collectively, this review provides an updated overview about the critical role of the p53/TAp73 pathway in the pathogenesis of NB, highlighting encouraging clues for the advance of alternative NB targeted therapies.
Collapse
|
12
|
Liu Z, Gao J, Gu R, Shi Y, Hu H, Liu J, Huang J, Zhong C, Zhou W, Yang Y, Gong C. Comprehensive Analysis of Transcriptomics and Genetic Alterations Identifies Potential Mechanisms Underlying Anthracycline Therapy Resistance in Breast Cancer. Biomolecules 2022; 12:biom12121834. [PMID: 36551262 PMCID: PMC9775906 DOI: 10.3390/biom12121834] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Anthracycline is a mainstay of treatment for breast cancer patients because of its antitumor activity. However, anthracycline resistance is a critical barrier in treating breast cancer. Thus, it is of great importance to uncover the molecular mechanisms underlying anthracycline resistance in breast cancer. Herein, we integrated transcriptome data, genetic alterations data, and clinical data of The Cancer Genome Atlas (TCGA) to identify the molecular mechanisms involved in anthracycline resistance in breast cancer. Two hundred and four upregulated genes and 1376 downregulated genes were characterized between the anthracycline-sensitive and anthracycline-resistant groups. It was found that drug resistance-associated genes such as ABCB5, CYP1A1, and CYP4Z1 were significantly upregulated in the anthracycline-resistant group. The gene set enrichment analysis (GSEA) suggested that the P53 signaling pathway, DNA replication, cysteine, and methionine metabolism pathways were associated with anthracycline sensitivity. Somatic TP53 mutation was a common genetic abnormality observed in the anthracycline-sensitive group, while CDH1 mutation was presented in the anthracycline-resistant group. Immune infiltration patterns were extremely different between the anthracycline-sensitive and anthracycline-resistant groups. Immune-associated chemokines and cytokines, immune regulators, and human leukocyte antigen genes were significantly upregulated in the anthracycline-sensitive group. These results reveal potential molecular mechanisms associated with anthracycline resistance.
Collapse
Affiliation(s)
- Zihao Liu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Department of Breast and Thyroid Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Jingbo Gao
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ran Gu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yu Shi
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Hong Hu
- Department of Breast and Thyroid Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Jianlan Liu
- Department of Pathology, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Jiefeng Huang
- Department of Breast and Thyroid Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Caineng Zhong
- Department of Breast and Thyroid Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Wenbin Zhou
- Department of Breast and Thyroid Surgery, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen 518020, China
| | - Yaping Yang
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Correspondence: (Y.Y.); or (C.G.)
| | - Chang Gong
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Correspondence: (Y.Y.); or (C.G.)
| |
Collapse
|
13
|
Wei Y, Wei C, Chen L, Liu N, Ou Q, Yin JC, Pang J, Fang Z, Wu X, Wang X, Mu D, Shao Y, Yu J, Yuan S. Genomic Correlates of Unfavorable Outcome in Locally Advanced Cervical Cancer Treated with Neoadjuvant Chemoradiation. Cancer Res Treat 2022; 54:1209-1218. [PMID: 35038823 PMCID: PMC9582489 DOI: 10.4143/crt.2021.963] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/13/2022] [Indexed: 11/21/2022] Open
Abstract
PURPOSE Neoadjuvant therapy modality can increase the operability rate and mitigate pathological risks in locally advanced cervical cancer, but treatment response varies widely. It remains unclear whether genetic alterations correlate with the response to neoadjuvant therapy and disease-free survival (DFS) in locally advanced cervical cancer. MATERIALS AND METHODS A total of 62 locally advanced cervical cancer (stage IB-IIA) patients who received neoadjuvant chemoradiation plus radical hysterectomy were retrospectively analyzed. Patients' tumor biopsy samples were comprehensively profiled using targeted next generation sequencing. Pathologic response to neoadjuvant treatment and DFS were evaluated against the association with genomic traits. RESULTS Genetic alterations of PIK3CA were most frequent (37%), comparable to that of Caucasian populations from The Cancer Genome Atlas. The mutation frequency of genes including TERT, POLD1, NOS2, and FGFR3 was significantly higher in Chinese patients whereas RPTOR, EGFR, and TP53 were underrepresented in comparison to Caucasians. Germline mutations were identified in 21% (13/62) of the cohort and more than half (57%) had mutations in DNA damage repair genes, including BRCA1/2, TP53 and PALB2. Importantly, high tumor mutation burden, TP53 polymorphism (rs1042522), and KEAP1 mutations were found to be associated with poor pathologic response to neoadjuvant chemoradiation treatment. KEAP1 mutations, PIK3CA-SOX2 co-amplification, TERC copy number gain, and TYMS polymorphism correlated with an increased risk of disease relapse. CONCLUSION We report the genomic profile of locally advanced cervical cancer patients and the distinction between Asian and Caucasian cohorts. Our findings highlight genomic traits associated with unfavorable neoadjuvant chemoradiation response and a higher risk of early disease recurrence.
Collapse
Affiliation(s)
- Yuchun Wei
- Cheeloo College of Medicine, Shandong University, Jinan,
China
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan,
China
| | - Chuqing Wei
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan,
China
| | - Liang Chen
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan,
China
| | - Ning Liu
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan,
China
| | - Qiuxiang Ou
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing,
China
| | - Jiani C. Yin
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing,
China
| | - Jiaohui Pang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing,
China
| | - Zhenhao Fang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing,
China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing,
China
| | - Xiaonan Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing,
China
| | - Dianbin Mu
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan,
China
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing,
China
- School of Public Health, Nanjing Medical University, Nanjing,
China
| | - Jinming Yu
- Cheeloo College of Medicine, Shandong University, Jinan,
China
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan,
China
| | - Shuanghu Yuan
- Cheeloo College of Medicine, Shandong University, Jinan,
China
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan,
China
| |
Collapse
|
14
|
Szpechcinski A, Szolkowska M, Winiarski S, Lechowicz U, Wisniewski P, Knetki-Wroblewska M. Targeted Next-Generation Sequencing of Thymic Epithelial Tumours Revealed Pathogenic Variants in KIT, ERBB2, KRAS, and TP53 in 30% of Thymic Carcinomas. Cancers (Basel) 2022; 14:3388. [PMID: 35884448 PMCID: PMC9324890 DOI: 10.3390/cancers14143388] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 01/27/2023] Open
Abstract
A better understanding of the molecular pathogenesis of thymic epithelial tumours (TETs) could revolutionise their treatment. We evaluated thymomas and thymic carcinomas by next-generation sequencing (NGS) of somatic or germline single nucleotide variants (SNVs) in genes commonly mutated in solid tumours. In total, 19 thymomas and 34 thymic carcinomas were analysed for nonsynonymous SNVs in 15 genes by targeted NGS (reference genome: hg19/GRCh37). Ten SNVs in TP53 (G154V, R158P, L194H, R267fs, R273C, R306 *, Q317 *), ERBB2 (V773M), KIT (L576P), and KRAS (Q61L) considered somatic and pathogenic/likely pathogenic were detected in 10 of 34 (29.4%) thymic carcinomas. No somatic SNVs confirmed as pathogenic/likely pathogenic were found in thymomas. Rare SNVs of uncertain or unknown functional and clinical significance, to our knowledge not reported previously in TETs, were found in ERBB2 (S703R), KIT (I690V), and FOXL2 (P157S) in 3 of 19 (16%) thymomas. The most frequent germline SNVs were TP53 P72R (94% TETs), ERBB2 I655V (40% TETs), and KIT M541L (9% TETs). No significant difference in median disease-free survival (DFS) was found between thymic carcinoma patients with and without pathogenic SNVs (p = 0.190); however, a trend toward a longer DFS was observed in the latter (16.0 vs. 30.0 months, respectively). In summary, NGS analysis of TETs revealed several SNVs in genes related to the p53, AKT, MAPK, and K-Ras signalling pathways. Thymic carcinomas showed greater genetic dysregulation than thymomas. The germline and rare SNVs of uncertain clinical significance reported in this study add to the number of known genetic alterations in TETs, thus extending our molecular understanding of these neoplasms. Druggable KIT alterations in thymic carcinomas have potential as therapeutic targets.
Collapse
Affiliation(s)
- Adam Szpechcinski
- Department of Genetics and Clinical Immunology, The Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland;
| | - Malgorzata Szolkowska
- Department of Pathology, The Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland
| | - Sebastian Winiarski
- Clinics of Thoracic Surgery, The Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland;
| | - Urszula Lechowicz
- Department of Genetics and Clinical Immunology, The Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland;
| | - Piotr Wisniewski
- Department of Pathology and Laboratory Medicine, The Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Magdalena Knetki-Wroblewska
- Department of Lung Cancer and Chest Tumours, The Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| |
Collapse
|
15
|
Malenge MM, Maaland AF, Repetto-Llamazares A, Middleton B, Nijland M, Visser L, Patzke S, Heyerdahl H, Kolstad A, Stokke T, Ree AH, Dahle J. Anti-CD37 radioimmunotherapy with 177Lu-NNV003 synergizes with the PARP inhibitor olaparib in treatment of non-Hodgkin’s lymphoma in vitro. PLoS One 2022; 17:e0267543. [PMID: 35486574 PMCID: PMC9053826 DOI: 10.1371/journal.pone.0267543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/11/2022] [Indexed: 11/18/2022] Open
Abstract
Background and purpose
PARP inhibitors have been shown to increase the efficacy of radiotherapy in preclinical models. Radioimmunotherapy results in selective radiation cytotoxicity of targeted tumour cells. Here we investigate the combined effect of anti-CD37 β-emitting 177Lu-NNV003 radioimmunotherapy and the PARP inhibitor olaparib, and gene expression profiles in CD37 positive non-Hodgkin’s lymphoma cell lines.
Materials and methods
The combined effect of 177Lu-NNV003 and olaparib was studied in seven cell lines using a fixed-ratio ray design, and combination index was calculated for each combination concentration. mRNA was extracted before and after treatment with the drug combination. After RNA-sequencing, hierarchical clustering was performed on basal gene expression profiles and on differentially expressed genes after combination treatment from baseline. Functional gene annotation analysis of significant differentially expressed genes after combination treatment was performed to identify enriched biological processes.
Results
The combination of olaparib and 177Lu-NNV003 was synergistic in four of seven cell lines, antagonistic in one and both synergistic and antagonistic (conditionally synergistic) in two, depending on the concentration ratio between olaparib and 177Lu-NNV003. Cells treated with the combination significantly overexpressed genes in the TP53 signalling pathway. However, cluster analysis did not identify gene clusters that correlate with the sensitivity of cells to single agent or combination treatment.
Conclusion
The cytotoxic effect of the combination of the PARP inhibitor olaparib and the β-emitting radioimmunoconjugate 177Lu-NNV003 was synergistic in the majority of tested lymphoma cell lines.
Collapse
Affiliation(s)
- Marion M. Malenge
- Nordic Nanovector ASA, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Astri Fjelde Maaland
- Nordic Nanovector ASA, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | - Marcel Nijland
- University Medical Center Groningen, Groningen, The Netherlands
| | - Lydia Visser
- University Medical Center Groningen, Groningen, The Netherlands
| | - Sebastian Patzke
- Nordic Nanovector ASA, Oslo, Norway
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | | | - Arne Kolstad
- Department of Oncology, Innlandet Sykehus, Lillehammer, Norway
| | - Trond Stokke
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Anne Hansen Ree
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| | | |
Collapse
|
16
|
Abstract
AIMS This study aims to investigate whether HRG gene C633T rs9898 and TP73 gene rs4648551 A > G polymorphisms have an effect on ovulation and response to the gonadotropin treatments. MATERIALS AND METHODS Blood samples were received from a total of 206 individuals (116 patients from whom good quality and optimal of numbers oocytes have not been able to be obtained at the IVF Center of Ondokuz Mayıs University, Faculty of Medicine and 90 controls). Genomic DNA was extracted by DNA isolation and SNP genotyping was performed by real-time qPCR method. RESULTS According to the results, a significant difference was observed between the patient and control groups in terms of the TP73 gene variant, however there was no significant difference regarding HRG gene polymorphism. CONCLUSIONS Our findings suggest that while AG genotype for TP73 could be a genetic marker for ovarian response, HRG gene C633T variation is not associated with ovarian response in our cohort. Further studies with larger study groups are required to investigate possible associations of these gene variants with ovarian response.
Collapse
Affiliation(s)
- Kadir Bakay
- Gynecology and Obstetrics, Ondokuz Mayis University Faculty of Medicine, Samsun, Turkey
| | - Ulas Coban
- Gynecology and Obstetrics, Ondokuz Mayis University Faculty of Medicine, Samsun, Turkey
| | - Mehmet Alper Arslan
- Medical Biology, Ondokuz Mayis University Faculty of Medicine, Samsun, Turkey
| | - Davut Guven
- Gynecology and Obstetrics, Ondokuz Mayis University Faculty of Medicine, Samsun, Turkey
| | - Sengul Tural
- Medical Biology, Ondokuz Mayis University Faculty of Medicine, Samsun, Turkey
| |
Collapse
|
17
|
van der Kamp MF, Halmos GB, Guryev V, Horvatovich PL, Schuuring E, van der Laan BFAM, van der Vegt B, Plaat BEC, Verhoeven CJ. Age-specific oncogenic pathways in head and neck squamous cell carcinoma - are elderly a different subcategory? Cell Oncol (Dordr) 2022; 45:1-18. [PMID: 35015241 DOI: 10.1007/s13402-021-00655-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In recent clinical practice, an increasing number of elderly patients suffering from head and neck squamous cell carcinoma (HNSCC) of unknown pathophysiology is observed. The majority of HNSCC patients can roughly be divided into three subcategories. First, a small group of young patients who present with variants of genomic aberrations and inheritable diseases like Fanconi anaemia. Second, an increasing population of HPV-related HNSCCs that are regarded as genomic stable tumours with a more favourable prognosis. Though HPV-related tumours used to be more common among younger males, a notable rise in the elderly population is observed. The third subcategory, that of HPV-negative tumours, has been shown to be more heterogeneous with involvement of a variety of oncogenic pathways related to lifestyle factors like smoking and alcohol consumption, often seen in middle-aged males. Some of these pathways could be related to age, such as TP53 alterations, EGFR activation, apoptotic pathway alterations and field cancerization. CONCLUSIONS In this narrative review, we provide an overview of established and newly discovered age-specific pathophysiological mechanisms underlying HNSCC. We propose a fourth subcategory of patients with a suspected different pathophysiology: elderly (HPV-negative) HNSCC patients without a history of tobacco and alcohol consumption. In this subcategory, carcinogenesis seems to be a multi-step process based on genomic instability, immunosenescence, cell cycle disruption and telomere shortening. To conclude, we discuss suggestions for future research to fill the knowledge gap about age-dependent HNSCC carcinogenesis.
Collapse
Affiliation(s)
- Martine Froukje van der Kamp
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands.
| | - Gyorgy Bela Halmos
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands
| | - Victor Guryev
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter Laszlo Horvatovich
- Department of Pharmacy, Analytical Biochemistry, University of Groningen, Groningen, The Netherlands
| | - Ed Schuuring
- Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Bert van der Vegt
- Department of Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Boudewijn Evert Christiaan Plaat
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands
| | - Cornelia Johanna Verhoeven
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB, Groningen, The Netherlands
| |
Collapse
|
18
|
Upregulation of p53 by tannic acid treatment suppresses the proliferation of human colorectal carcinoma. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2021; 71:587-602. [PMID: 36651555 DOI: 10.2478/acph-2021-0036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 01/19/2023]
Abstract
The present study's objective is to clarify the molecular mechanisms of tannic acid effects on the viability of human colorectal carcinoma (CRC). Tannic acid is stable for up to 48 h and is localized in both cytoplasm and nucleus. It dose-dependently inhibited the viability of CRC cell lines; SW-620 and HT-29 with IC 50 values of 7.2 ± 0.8 and 37.6 ± 1.4 µmol L-1. Besides, metastatic, invasive, and colony formation properties of CRC cells were significantly inhibited following the tannic acid treatment (p < 0.001). Tannic acid has been found to modulate enzyme, protein, and gene expressions of NQO1 in different levels and the upregulation of protein/gene expressions of p53 (p < 0.001), which leads the cells to trigger apoptosis. In conclusion, the present in vitro study may supply a significant background for in vivo studies in which the molecular mechanisms of antioxidant and chemopreventive activities of tannic acid will completely clarify.
Collapse
|
19
|
Dong Y, Wang J, Ji W, Zheng M, Wang P, Liu L, Li S. Preclinical Application of Conditional Reprogramming Culture System for Laryngeal and Hypopharyngeal Carcinoma. Front Cell Dev Biol 2021; 9:744969. [PMID: 34778255 PMCID: PMC8585768 DOI: 10.3389/fcell.2021.744969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Management of laryngeal and hypopharyngeal squamous cell carcinoma (LHSCC) remains highly challenging due to highly variable therapeutic responses. By establishing an in vitro model for LHSCC based on conditional reprogramming (CR), a cell-culture technique, we aim to investigate its potential value on personalized cancer therapies. Herein, a panel of 28 human LHSCC CR cells were established from 50 tumor tissues using the CR method. They retained tumorigenic potential upon xenotransplantation and recapitulated molecular characteristics of LHSCC. Differential responses to anticancer drugs and radiotherapy were detected in vitro. CR cells could be transformed to xenograft and organoid, and they shared comparable drug responses. The clinical drug responses were consistent with in vitro drug responses. Collectively, the patient-derived CR cell model could promisingly be utilized in clinical decision-making and assisted in the selection of personalized therapies for LHSCC.
Collapse
Affiliation(s)
- Yanbo Dong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jian Wang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Wei Ji
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mengzhu Zheng
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Peng Wang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Liangfa Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
20
|
Porpaczy E, Wohlfarth P, Königsbrügge O, Rabitsch W, Skrabs C, Staber P, Worel N, Müllauer L, Simonitsch-Klupp I, Kornauth C, Rohrbeck J, Jaeger U, Schiefer AI. Influence of TP53 Mutation on Survival of Diffuse Large B-Cell Lymphoma in the CAR T-Cell Era. Cancers (Basel) 2021; 13:cancers13225592. [PMID: 34830747 PMCID: PMC8616128 DOI: 10.3390/cancers13225592] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/28/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary The genetic landscape of diffuse large B-cell lymphoma (DLBCL) is heterogenous. So far, detailed studies about TP53 mutations in DLBCL treated with anti-CD19 chimeric antigen receptor T-cell (CAR T cells) therapy are still missing. Chemotherapy resistance is one of the challenges in TP53 mutated tumors. New immunomodulatory agents, such as different inhibitors or CAR T cells, have shown durable responses in refractory/relapsed DLBCL in recent years. Although our CAR T cell treated cohort was small, we aimed to investigate the influence of TP53 mutations on overall survival of patients treated with CAR T cells compared to DLBCL patients without CAR T-cell therapy. Identification of risk factors for treatment failure may aid in choosing the most promising treatment in every setting. Abstract Refractory/relapsed diffuse large B-cell lymphoma (DLBCL) is associated with poor outcome. The clinical behavior and genetic landscape of DLBCL is heterogeneous and still not fully understood. TP53 mutations in DLBCL have been identified as markers of poor prognosis and are often associated with therapeutic resistance. Chimeric antigen receptor T-cell therapy is an innovative therapeutic concept and represents a game-changing therapeutic option by supporting the patient’s own immune system to kill the tumor cells. We investigated the impact of TP53 mutations on the overall survival of refractory/relapsed DLBCL patients treated with comparable numbers of therapy lines. The minimum number of therapy lines was 2 (median 4), including either anti-CD19 CAR T-cell therapy or conventional salvage therapy. A total of 170 patients with DLBCL and high-grade B-cell lymphoma with MYC, BCL2, and/or BCL6 rearrangements (DHL/THL), diagnosed and treated in our hospital between 2000 and 2021, were included. Twenty-nine of them received CAR T-cell therapy. TP53 mutations were found in 10/29 (35%) and 31/141 (22%) of patients in the CAR T-cell and conventional groups, respectively. Among the 141 patients not treated with CAR T cells, TP53 mutation was an independent prognostic factor for overall survival (OS) (median 12 months with TP53 vs. not reached without TP53 mutation, p < 0.005), but in the CAR T cell treated group, this significance could not be shown (median OS 30 vs. 120 months, p = 0.263). The findings from this monocentric retrospective study indicate that TP53 mutation status does not seem to affect outcomes in DLBCL patients treated with CAR T-cell therapy. Detailed evaluation in large cohorts is warranted.
Collapse
Affiliation(s)
- Edit Porpaczy
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (E.P.); (O.K.); (C.S.); (P.S.); (U.J.)
| | - Philipp Wohlfarth
- Department of Internal Medicine I, Hematopoietic Stem Cell Transplantation Unit, Medical University of Vienna, 1090 Vienna, Austria; (P.W.); (W.R.)
| | - Oliver Königsbrügge
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (E.P.); (O.K.); (C.S.); (P.S.); (U.J.)
| | - Werner Rabitsch
- Department of Internal Medicine I, Hematopoietic Stem Cell Transplantation Unit, Medical University of Vienna, 1090 Vienna, Austria; (P.W.); (W.R.)
| | - Cathrin Skrabs
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (E.P.); (O.K.); (C.S.); (P.S.); (U.J.)
| | - Philipp Staber
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (E.P.); (O.K.); (C.S.); (P.S.); (U.J.)
| | - Nina Worel
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (L.M.); (I.S.-K.); (C.K.); (J.R.)
| | - Ingrid Simonitsch-Klupp
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (L.M.); (I.S.-K.); (C.K.); (J.R.)
| | - Christoph Kornauth
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (L.M.); (I.S.-K.); (C.K.); (J.R.)
| | - Johannes Rohrbeck
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (L.M.); (I.S.-K.); (C.K.); (J.R.)
| | - Ulrich Jaeger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (E.P.); (O.K.); (C.S.); (P.S.); (U.J.)
| | - Ana-Iris Schiefer
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (L.M.); (I.S.-K.); (C.K.); (J.R.)
- Correspondence:
| |
Collapse
|
21
|
Noor H, Briggs NE, McDonald KL, Holst J, Vittorio O. TP53 Mutation Is a Prognostic Factor in Lower Grade Glioma and May Influence Chemotherapy Efficacy. Cancers (Basel) 2021; 13:5362. [PMID: 34771529 PMCID: PMC8582451 DOI: 10.3390/cancers13215362] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/17/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Identification of prognostic biomarkers in cancers is a crucial step to improve overall survival (OS). Although mutations in tumour protein 53 (TP53) is prevalent in astrocytoma, the prognostic effects of TP53 mutation are unclear. METHODS In this retrospective study, we sequenced TP53 exons 1 to 10 in a cohort of 102 lower-grade glioma (LGG) subtypes and determined the prognostic effects of TP53 mutation in astrocytoma and oligodendroglioma. Publicly available datasets were analysed to confirm the findings. RESULTS In astrocytoma, mutations in TP53 codon 273 were associated with a significantly increased OS compared to the TP53 wild-type (HR (95% CI): 0.169 (0.036-0.766), p = 0.021). Public datasets confirmed these findings. TP53 codon 273 mutant astrocytomas were significantly more chemosensitive than TP53 wild-type astrocytomas (HR (95% CI): 0.344 (0.13-0.88), p = 0.0148). Post-chemotherapy, a significant correlation between TP53 and YAP1 mRNA was found (p = 0.01). In O (6)-methylguanine methyltransferase (MGMT) unmethylated chemotherapy-treated astrocytoma, both TP53 codon 273 and YAP1 mRNA were significant prognostic markers. In oligodendroglioma, TP53 mutations were associated with significantly decreased OS. CONCLUSIONS Based on these findings, we propose that certain TP53 mutant astrocytomas are chemosensitive through the involvement of YAP1, and we outline a potential mechanism. Thus, TP53 mutations may be key drivers of astrocytoma therapeutic efficacy and influence survival outcomes.
Collapse
Affiliation(s)
- Humaira Noor
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, NSW 2031, Australia;
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Randwick, NSW 2031, Australia;
| | - Nancy E. Briggs
- Stats Central, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2031, Australia;
| | - Kerrie L. McDonald
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, NSW 2031, Australia;
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Randwick, NSW 2031, Australia;
| | - Jeff Holst
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Randwick, NSW 2031, Australia;
- Translational Cancer Metabolism Laboratory, School of Medical Sciences, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2031, Australia
| | - Orazio Vittorio
- School of Women’s & Children’s Health, UNSW Medicine, University of NSW, Randwick, NSW 2031, Australia;
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, NSW 2031, Australia
| |
Collapse
|
22
|
TP53 in Acute Myeloid Leukemia: Molecular Aspects and Patterns of Mutation. Int J Mol Sci 2021; 22:ijms221910782. [PMID: 34639121 PMCID: PMC8509740 DOI: 10.3390/ijms221910782] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 01/10/2023] Open
Abstract
Mutation of the tumor suppressor gene, TP53, is associated with abysmal survival outcomes in acute myeloid leukemia (AML). Although it is the most commonly mutated gene in cancer, its occurrence is observed in only 5–10% of de novo AML, and in 30% of therapy related AML (t-AML). TP53 mutation serves as a prognostic marker of poor response to standard-of-care chemotherapy, particularly in t-AML and AML with complex cytogenetics. In light of a poor response to traditional chemotherapy and only a modest improvement in outcome with hypomethylation-based interventions, allogenic stem cell transplant is routinely recommended in these cases, albeit with a response that is often short lived. Despite being frequently mutated across the cancer spectrum, progress and enthusiasm for the development of p53 targeted therapeutic interventions is lacking and to date there is no approved drug that mitigates the effects of TP53 mutation. There is a mounting body of evidence indicating that p53 mutants differ in functionality and form from typical AML cases and subsequently display inconsistent responses to therapy at the cellular level. Understanding this pathobiological activity is imperative to the development of effective therapeutic strategies. This review aims to provide a comprehensive understanding of the effects of TP53 on the hematopoietic system, to describe its varying degree of functionality in tumor suppression, and to illustrate the need for the adoption of personalized therapeutic strategies to target distinct classes of the p53 mutation in AML management.
Collapse
|
23
|
Wiegmans AP, Ward A, Ivanova E, Duijf PHG, Adams MN, Najib IM, Van Oosterhout R, Sadowski MC, Kelly G, Morrical SW, O'Byrne K, Lee JS, Richard DJ. Genome instability and pressure on non-homologous end joining drives chemotherapy resistance via a DNA repair crisis switch in triple negative breast cancer. NAR Cancer 2021; 3:zcab022. [PMID: 34316709 PMCID: PMC8210242 DOI: 10.1093/narcan/zcab022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 05/09/2021] [Accepted: 05/24/2021] [Indexed: 11/29/2022] Open
Abstract
Chemotherapy is used as a standard-of-care against cancers that display high levels of inherent genome instability. Chemotherapy induces DNA damage and intensifies pressure on the DNA repair pathways that can lead to deregulation. There is an urgent clinical need to be able to track the emergence of DNA repair driven chemotherapy resistance and tailor patient staging appropriately. There have been numerous studies into chemoresistance but to date no study has elucidated in detail the roles of the key DNA repair components in resistance associated with the frontline clinical combination of anthracyclines and taxanes together. In this study, we hypothesized that the emergence of chemotherapy resistance in triple negative breast cancer was driven by changes in functional signaling in the DNA repair pathways. We identified that consistent pressure on the non-homologous end joining pathway in the presence of genome instability causes failure of the key kinase DNA-PK, loss of p53 and compensation by p73. In-turn a switch to reliance on the homologous recombination pathway and RAD51 recombinase occurred to repair residual double strand DNA breaks. Further we demonstrate that RAD51 is an actionable target for resensitization to chemotherapy in resistant cells with a matched gene expression profile of resistance highlighted by homologous recombination in clinical samples.
Collapse
Affiliation(s)
- Adrian P Wiegmans
- Queensland University of Technology (QUT), Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Woolloongabba QLD 4121, Australia
| | - Ambber Ward
- School of Medicine, University of Queensland, St Lucia, QLD Australia
| | - Ekaterina Ivanova
- Queensland University of Technology (QUT), Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Woolloongabba QLD 4121, Australia
| | - Pascal H G Duijf
- Queensland University of Technology (QUT), Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Woolloongabba QLD 4121, Australia
| | - Mark N Adams
- Queensland University of Technology (QUT), Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Woolloongabba QLD 4121, Australia
| | - Idris Mohd Najib
- Queensland University of Technology (QUT), Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Woolloongabba QLD 4121, Australia
| | - Romy Van Oosterhout
- Tumor Microenvironment Laboratory, QIMR Berghofer, Herston Rd, Herston, QLD 4006, Australia
| | - Martin C Sadowski
- Queensland University of Technology (QUT), Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Woolloongabba QLD 4121, Australia
| | - Greg Kelly
- Tumor Microenvironment Laboratory, QIMR Berghofer, Herston Rd, Herston, QLD 4006, Australia
| | - Scott W Morrical
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Ken O'Byrne
- Queensland University of Technology (QUT), Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Woolloongabba QLD 4121, Australia
| | - Jason S Lee
- School of Medicine, University of Queensland, St Lucia, QLD Australia
| | - Derek J Richard
- Queensland University of Technology (QUT), Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Translational Research Institute, Woolloongabba QLD 4121, Australia
| |
Collapse
|
24
|
Genetic associations of TP53 codon Pro72Arg polymorphism (rs1042522) in coronary artery disease: A meta-analysis of candidate genetic mutants. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
25
|
Barrera G, Cucci MA, Grattarola M, Dianzani C, Muzio G, Pizzimenti S. Control of Oxidative Stress in Cancer Chemoresistance: Spotlight on Nrf2 Role. Antioxidants (Basel) 2021; 10:antiox10040510. [PMID: 33805928 PMCID: PMC8064392 DOI: 10.3390/antiox10040510] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
Chemoresistance represents the main obstacle to cancer treatment with both conventional and targeted therapy. Beyond specific molecular alterations, which can lead to targeted therapy, metabolic remodeling, including the control of redox status, plays an important role in cancer cell survival following therapy. Although cancer cells generally have a high basal reactive oxygen species (ROS) level, which makes them more susceptible than normal cells to a further increase of ROS, chemoresistant cancer cells become highly adapted to intrinsic or drug-induced oxidative stress by upregulating their antioxidant systems. The antioxidant response is principally mediated by the transcription factor Nrf2, which has been considered the master regulator of antioxidant and cytoprotective genes. Nrf2 expression is often increased in several types of chemoresistant cancer cells, and its expression is mediated by diverse mechanisms. In addition to Nrf2, other transcription factors and transcriptional coactivators can participate to maintain the high antioxidant levels in chemo and radio-resistant cancer cells. The control of expression and function of these molecules has been recently deepened to identify which of these could be used as a new therapeutic target in the treatment of tumors resistant to conventional therapy. In this review, we report the more recent advances in the study of Nrf2 regulation in chemoresistant cancers and the role played by other transcription factors and transcriptional coactivators in the control of antioxidant responses in chemoresistant cancer cells.
Collapse
Affiliation(s)
- Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; (M.A.C.); (M.G.); (G.M.); (S.P.)
- Correspondence:
| | - Marie Angele Cucci
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; (M.A.C.); (M.G.); (G.M.); (S.P.)
| | - Margherita Grattarola
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; (M.A.C.); (M.G.); (G.M.); (S.P.)
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 11, 10125 Turin, Italy;
| | - Giuliana Muzio
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; (M.A.C.); (M.G.); (G.M.); (S.P.)
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Turin, Corso Raffaello 30, 10125 Turin, Italy; (M.A.C.); (M.G.); (G.M.); (S.P.)
| |
Collapse
|
26
|
Li H, Chen L, Tong X, Dai H, Shi T, Cheng X, Sun M, Chen K, Wei Q, Wang M. Functional genetic variants of CTNNBIP1 predict platinum treatment response of Chinese epithelial ovarian cancer patients. J Cancer 2020; 11:6850-6860. [PMID: 33123276 PMCID: PMC7592014 DOI: 10.7150/jca.48218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/15/2020] [Indexed: 12/29/2022] Open
Abstract
Chemotherapy resistance remains a blockade for successful treatment and longer overall survival of patients with epithelial ovarian cancer (EOC). CTNNBIP1 is an inhibitor of β-catenin that is a chemotherapeutic target for EOC treatment. In the present study, we investigated associations between single nucleotide polymorphisms (SNPs) of CTNNBIP1 and platinum treatment response of Han Chinese EOC patients and subsequently performed functional prediction and validation of the resultant SNPs. We found that CTNNBIP1 rs935072 AT/TT variant genotypes were associated with platinum treatment response in the multivariate logistic regression analysis of EOC patients. Specifically, the CTNNBIP1 rs935072 AT/TT genotypes were associated with a decreased risk of developing chemoresistance ([adjusted odds ratio (OR)] = 0.89, 95% confidence interval (CI) = 0.82-0.97 and P=0.010), compared with the AA genotype. Further experiments showed that the underlying mechanism for the CTNNBIP1 rs935072 A>T change in chemotherapy treatment response resulted from a lower binding affinity of miR-27a-3p, thereby leading to up-regulation of the CTNNBIP1 expression. We further found that overexpression of CTNNBIP1 sensitized ovarian cancer cells to platinum treatment. Thus, the present study provides evidence that functional variants of CTNNBIP1 may regulate the expression of CTNNBIP1, a possible mechanism affecting platinum treatment response of EOC patients.
Collapse
Affiliation(s)
- Haoran Li
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lihua Chen
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoxia Tong
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongji Dai
- Department of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Tingyan Shi
- Ovarian Cancer Program, Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, Fudan University Zhongshan Hospital, Shanghai, China
| | - Xi Cheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Menghong Sun
- Department of Pathology, Tissue Bank, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qingyi Wei
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Mengyun Wang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Intronic TP53 Polymorphisms Are Associated with Increased Δ133TP53 Transcript, Immune Infiltration and Cancer Risk. Cancers (Basel) 2020; 12:cancers12092472. [PMID: 32882831 PMCID: PMC7563340 DOI: 10.3390/cancers12092472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022] Open
Abstract
We investigated the influence of selected TP53 SNPs in exon 4 and intron 4 on cancer risk, clinicopathological features and expression of TP53 isoforms. The intron 4 SNPs were significantly over-represented in cohorts of mixed cancers compared to three ethnically matched controls, suggesting they confer increased cancer risk. Further analysis showed that heterozygosity at rs1042522(GC) and either of the two intronic SNPs rs9895829(TC) and rs2909430(AG) confer a 2.34-5.35-fold greater risk of developing cancer. These SNP combinations were found to be associated with shorter patient survival for glioblastoma and prostate cancer. Additionally, these SNPs were associated with tumor-promoting inflammation as evidenced by high levels of infiltrating immune cells and expression of the Δ133TP53 and TP53β transcripts. We propose that these SNP combinations allow increased expression of the Δ133p53 isoforms to promote the recruitment of immune cells that create an immunosuppressive environment leading to cancer progression.
Collapse
|
28
|
Sambyal V, Kaur S, Manjari M, Uppal MS, Singh NR, Sudan M, Guleria K. Linkage disequilibrium and haplotypes of five TP53 polymorphisms in oesophageal cancer patients. J Genet 2020. [DOI: 10.1007/s12041-020-01224-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Nassar A, Abouelhoda M, Mansour O, Loutfy SA, Hafez MM, Gomaa M, Bahnassy A, El-Din Youssef AS, Lotfy MM, Ismail H, Ahmed OS, Abou-Bakr AAE, Zekri ARN. Targeted next generation sequencing identifies somatic mutations in a cohort of Egyptian breast cancer patients. J Adv Res 2020; 24:149-157. [PMID: 32322420 PMCID: PMC7167517 DOI: 10.1016/j.jare.2020.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/17/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer (BC) incidence is progressively increasing in Egypt. However, there is insufficient knowledge of the acquired somatic mutations in Egyptian BC patients which limit our understanding of its progression. To the best of our knowledge, this is the first Egyptian cohort to sequence a multiple-gene panel of cancer related genes on BC patients. Four hundred and nine cancer related genes were sequenced in 46 fresh breast tumors of Egyptian BC patients to identify somatic mutations and their frequencies. TP53 and PIK3CA were the most top two frequently mutated genes. We detected 15 different somatic mutations in TP53 and 8 different ones in PIK3CA, each in 27 samples (58.7%). According to Clinvar database; we found 19 pathogenic somatic mutations: 7 in Tp53, 5 in PIK3CA, and single variants of VHL, STK11, AKT1, KRAS, IDH2, PTEN and ERBB2. We also identified 5 variants with uncertain significance (4 in TP53 and 1 in CEBPA) and 4 variants with conflicting interpretations of pathogenicity (2 in TP53 and 1 in each of APC and JAK3). Moreover, one drug response variant (p.P72R) in TP53 was detected in 8 samples. Furthermore, four novel variants were identified in JAK2, MTOR, KIT and EPHB. Further analysis, by Ingenuity Variant Analysis software (IVA), showed that PI3K/AKT signaling is altered in greater than 50% of Egyptian BC patients which implicates PI3K/AKT signaling as a therapeutic target. In this cohort, we shed the light on the most frequently detected somatic mutations and the most altered pathway in Egyptian BC patients.
Collapse
Affiliation(s)
- Auhood Nassar
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Osman Mansour
- Medical Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Samah A. Loutfy
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed M. Hafez
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - M. Gomaa
- Radiology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Abeer Bahnassy
- Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Mai M. Lotfy
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Hoda Ismail
- Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Ola S. Ahmed
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Abdel-Rahman N. Zekri
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
30
|
Lopes EA, Gomes S, Saraiva L, Santos MM. Small Molecules Targeting Mutant P53: A Promising Approach for Cancer Treatment. Curr Med Chem 2020; 26:7323-7336. [DOI: 10.2174/0929867325666181116124308] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/11/2018] [Accepted: 10/25/2018] [Indexed: 12/17/2022]
Abstract
:
More than half of all human tumors express mutant forms of p53, with the ovary,
lung, pancreas, and colorectal cancers among the tumor types that display the highest prevalence
of p53 mutations. In addition, the expression of mutant forms of p53 in tumors is associated
with poor prognosis due to increased chemoresistance and invasiveness. Therefore, the
pharmacological restoration of wild-type-like activity to mutant p53 arises as a promising therapeutic
strategy against cancer. This review is focused on the most relevant mutant p53 small
molecule reactivators described to date. Despite some of them have entered into clinical trials,
none has reached the clinic, which emphasizes that new pharmacological alternatives, particularly
with higher selectivity and lower adverse toxic side effects, are still required.
Collapse
Affiliation(s)
- Elizabeth A. Lopes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Gomes
- LAQV-REQUIMTE, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Lucília Saraiva
- LAQV-REQUIMTE, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Maria M.M. Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
31
|
Miller JJ, Blanchet A, Orvain C, Nouchikian L, Reviriot Y, Clarke RM, Martelino D, Wilson D, Gaiddon C, Storr T. Bifunctional ligand design for modulating mutant p53 aggregation in cancer. Chem Sci 2019; 10:10802-10814. [PMID: 32055386 PMCID: PMC7006507 DOI: 10.1039/c9sc04151f] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/06/2019] [Indexed: 12/19/2022] Open
Abstract
Protein misfolding and aggregation contributes to the development of a wide range of diseases. In cancer, over 50% of diagnoses are attributed to p53 malfunction due to missense mutations, many of which result in protein misfolding and accelerated aggregation. p53 mutations also frequently result in alteration or loss of zinc at the DNA-binding site, which increases aggregation via nucleation with zinc-bound p53. Herein, we designed two novel bifunctional ligands, LI and LH , to modulate mutant p53 aggregation and restore zinc binding using a metallochaperone approach. Interestingly, only the incorporation of iodine function in LI resulted in modulation of mutant p53 aggregation, both in recombinant and cellular environments. Native mass spectrometry shows a protein-ligand interaction for LI , as opposed to LH , which is hypothesized to lead to the distinct difference in the p53 aggregation profile for the two ligands. Incorporation of a di-2-picolylamine binding unit into the ligand design provided efficient intracellular zinc uptake, resulting in metallochaperone capability for both LI and LH . The ability of LI to reduce mutant p53 aggregation results in increased restoration of p53 transcriptional function and mediates both caspase-dependent and -independent cell death pathways. We further demonstrate that LI exhibits minimal toxicity in non-cancerous organoids, and that it is well tolerated in mice. These results demonstrate that iodination of our ligand framework restores p53 function by interacting with and inhibiting mutant p53 aggregation and highlights LI as a suitable candidate for comprehensive in vivo anticancer preclinical evaluations.
Collapse
Affiliation(s)
- Jessica J Miller
- Department of Chemistry , Simon Fraser University , Burnaby , British Columbia V5A 1S6 , Canada .
| | - Anaïs Blanchet
- Inserm UMR_S 1113 , Université de Strasbourg , Molecular Mechanisms of Stress Response and Pathologies , Strasbourg , France .
| | - Christophe Orvain
- Inserm UMR_S 1113 , Université de Strasbourg , Molecular Mechanisms of Stress Response and Pathologies , Strasbourg , France .
| | - Lucienne Nouchikian
- Chemistry Department , York University , 6 Thompson Road , Toronto , Ontario M3J 1L3 , Canada
| | - Yasmin Reviriot
- Department of Chemistry , Simon Fraser University , Burnaby , British Columbia V5A 1S6 , Canada .
| | - Ryan M Clarke
- Department of Chemistry , Simon Fraser University , Burnaby , British Columbia V5A 1S6 , Canada .
| | - Diego Martelino
- Department of Chemistry , Simon Fraser University , Burnaby , British Columbia V5A 1S6 , Canada .
| | - Derek Wilson
- Chemistry Department , York University , 6 Thompson Road , Toronto , Ontario M3J 1L3 , Canada
| | - Christian Gaiddon
- Inserm UMR_S 1113 , Université de Strasbourg , Molecular Mechanisms of Stress Response and Pathologies , Strasbourg , France .
| | - Tim Storr
- Department of Chemistry , Simon Fraser University , Burnaby , British Columbia V5A 1S6 , Canada .
| |
Collapse
|
32
|
Benyelles M, Episkopou H, O'Donohue M, Kermasson L, Frange P, Poulain F, Burcu Belen F, Polat M, Bole‐Feysot C, Langa‐Vives F, Gleizes P, de Villartay J, Callebaut I, Decottignies A, Revy P. Impaired telomere integrity and rRNA biogenesis in PARN-deficient patients and knock-out models. EMBO Mol Med 2019; 11:e10201. [PMID: 31273937 PMCID: PMC6609912 DOI: 10.15252/emmm.201810201] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/24/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
PARN, poly(A)-specific ribonuclease, regulates the turnover of mRNAs and the maturation and stabilization of the hTR RNA component of telomerase. Biallelic PARN mutations were associated with Høyeraal-Hreidarsson (HH) syndrome, a rare telomere biology disorder that, because of its severity, is likely not exclusively due to hTR down-regulation. Whether PARN deficiency was affecting the expression of telomere-related genes was still unclear. Using cells from two unrelated HH individuals carrying novel PARN mutations and a human PARN knock-out (KO) cell line with inducible PARN complementation, we found that PARN deficiency affects both telomere length and stability and down-regulates the expression of TRF1, TRF2, TPP1, RAP1, and POT1 shelterin transcripts. Down-regulation of dyskerin-encoding DKC1 mRNA was also observed and found to result from p53 activation in PARN-deficient cells. We further showed that PARN deficiency compromises ribosomal RNA biogenesis in patients' fibroblasts and cells from heterozygous Parn KO mice. Homozygous Parn KO however resulted in early embryonic lethality that was not overcome by p53 KO. Our results refine our knowledge on the pleiotropic cellular consequences of PARN deficiency.
Collapse
Affiliation(s)
- Maname Benyelles
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| | | | - Marie‐Françoise O'Donohue
- Laboratoire de Biologie Moléculaire EucaryoteCentre de Biologie Intégrative (CBI)CNRS, UPSUniversité de ToulouseToulouseFrance
| | - Laëtitia Kermasson
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| | - Pierre Frange
- EA 7327, Université Paris Descartes, Sorbonne Paris‐CitéParisFrance
- Laboratoire de Microbiologie clinique & Unité d'ImmunologieHématologie et Rhumatologie PédiatriquesAP‐HP, Hôpital Necker, Enfants MaladesParisFrance
| | - Florian Poulain
- de Duve InstituteUniversité catholique de LouvainBrusselsBelgium
| | - Fatma Burcu Belen
- Pediatric HematologyFaculty of MedicineBaskent UniversityAnkaraTurkey
| | - Meltem Polat
- Pediatric Infectious DiseasesDepartment of Pediatric Infectious DiseasesPamukkale University Medical FacultyDenizliTurkey
| | - Christine Bole‐Feysot
- INSERM, UMR 1163Genomics platform, Imagine InstituteParis Descartes–Sorbonne Paris Cité UniversityParisFrance
- Genomic Core FacilityImagine Institute‐Structure Fédérative de Recherche NeckerINSERM U1163ParisFrance
| | | | - Pierre‐Emmanuel Gleizes
- Laboratoire de Biologie Moléculaire EucaryoteCentre de Biologie Intégrative (CBI)CNRS, UPSUniversité de ToulouseToulouseFrance
| | - Jean‐Pierre de Villartay
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| | - Isabelle Callebaut
- Muséum National d'Histoire NaturelleUMR CNRS 7590Institut de Minéralogiede Physique des Matériaux et de Cosmochimie, IMPMCSorbonne UniversitéParisFrance
| | | | - Patrick Revy
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| |
Collapse
|
33
|
Yi Y, Zhang W, Yi J, Xiao ZX. Role of p53 Family Proteins in Metformin Anti-Cancer Activities. J Cancer 2019; 10:2434-2442. [PMID: 31258748 PMCID: PMC6584340 DOI: 10.7150/jca.30659] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 04/23/2019] [Indexed: 12/13/2022] Open
Abstract
Metformin has been used as therapy for type 2 diabetes for many years. Clinical and basic evidence as indicated that metformin has anti-cancer activities. It has been well-established that metformin activates AMP-activated protein kinase (AMPK), which in turn regulates energy homeostasis. However, the mechanistic aspects of metformin anti-cancer activity remain elusive. p53 family proteins, including p53, p63 and p73, have diverse biological functions, including regulation of cell growth, survival, development, senescence and aging. In this review, we highlight the evidence and mechanisms by which metformin inhibits cancer cell survival and tumor growth. We also aimed to discuss the role of p53 family proteins in metformin-mediated suppression of cancer growth and survival.
Collapse
|
34
|
Mukhopadhyay UK, Oturkar CC, Adams C, Wickramasekera N, Bansal S, Medisetty R, Miller A, Swetzig WM, Silwal-Pandit L, Børresen-Dale AL, Creighton CJ, Park JH, Konduri SD, Mukhopadhyay A, Caradori A, Omilian A, Bshara W, Kaipparettu BA, Das GM. TP53 Status as a Determinant of Pro- vs Anti-Tumorigenic Effects of Estrogen Receptor-Beta in Breast Cancer. J Natl Cancer Inst 2019; 111:1202-1215. [PMID: 30990221 PMCID: PMC6855950 DOI: 10.1093/jnci/djz051] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 12/28/2018] [Accepted: 04/01/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Anti-tumorigenic vs pro-tumorigenic roles of estrogen receptor-beta (ESR2) in breast cancer remain unsettled. We investigated the potential of TP53 status to be a determinant of the bi-faceted role of ESR2 and associated therapeutic implications for triple negative breast cancer (TNBC). METHODS ESR2-TP53 interaction was analyzed with multiple assays including the in situ proximity ligation assay. Transcriptional effects on TP53-target genes and cell proliferation in response to knocking down or overexpressing ESR2 were determined. Patient survival according to ESR2 expression levels and TP53 mutation status was analyzed in the basal-like TNBC subgroup in the Molecular Taxonomy of Breast Cancer International Consortium (n = 308) and Roswell Park Comprehensive Cancer Center (n = 46) patient cohorts by univariate Cox regression and log-rank test. All statistical tests are two-sided. RESULTS ESR2 interaction with wild-type and mutant TP53 caused pro-proliferative and anti-proliferative effects, respectively. Depleting ESR2 in cells expressing wild-type TP53 resulted in increased expression of TP53-target genes CDKN1A (control group mean [SD] = 1 [0.13] vs ESR2 depletion group mean [SD] = 2.08 [0.24], P = .003) and BBC3 (control group mean [SD] = 1 [0.06] vs ESR2 depleted group mean [SD] = 1.92 [0.25], P = .003); however, expression of CDKN1A (control group mean [SD] = 1 [0.21] vs ESR2 depleted group mean [SD] = 0.56 [0.12], P = .02) and BBC3 (control group mean [SD] = 1 [0.03] vs ESR2 depleted group mean [SD] = 0.55 [0.09], P = .008) was decreased in cells expressing mutant TP53. Overexpressing ESR2 had opposite effects. Tamoxifen increased ESR2-mutant TP53 interaction, leading to reactivation of TP73 and apoptosis. High levels of ESR2 expression in mutant TP53-expressing basal-like tumors is associated with better prognosis (Molecular Taxonomy of Breast Cancer International Consortium cohort: log-rank P = .001; hazard ratio = 0.26, 95% confidence interval = 0.08 to 0.84, univariate Cox P = .02). CONCLUSIONS TP53 status is a determinant of the functional duality of ESR2. Our study suggests that ESR2-mutant TP53 combination prognosticates survival in TNBC revealing a novel strategy to stratify TNBC for therapeutic intervention potentially by repurposing tamoxifen.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Gokul M Das
- Correspondence to: Gokul M. Das, PhD, Department of Pharmacology and Therapeutics, Center for Genetics and Pharmacology, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 (e-mail: )
| |
Collapse
|
35
|
Assoun S, Theou-Anton N, Nguenang M, Cazes A, Danel C, Abbar B, Pluvy J, Gounant V, Khalil A, Namour C, Brosseau S, Zalcman G. Association of TP53 mutations with response and longer survival under immune checkpoint inhibitors in advanced non-small-cell lung cancer. Lung Cancer 2019; 132:65-71. [PMID: 31097096 DOI: 10.1016/j.lungcan.2019.04.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 03/30/2019] [Accepted: 04/06/2019] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Tumor mutational burden (TMB) correlates with response to immune checkpoint inhibitors (ICI) in advanced non-small-cell lung cancer (aNSCLC). We hypothesized that TP53 mutations could reflect TMB and be associated with ICI benefit. METHODS TP53 mutations were assessed by next-generation sequencing in aNSCLC patients treated with programmed death-1 (PD-1) blockers. Clinical data, tumor programmed death ligand-1 (PD-L1) expression, and KRAS mutational status were collected. The primary endpoint was overall survival (OS). RESULTS In total, 72 patients (median [interquartile range] age: 61 [33-83] years) were included; 52 (72%) were male; 39 (54%) had performance status 0-1; 53 (74%) had adenocarcinoma; 20 (28%) received first-line ICI, 52 (72%) second line or more. In 65 patients with available data, 36 (55%) expressed PD-L1 in ≥50% of tumor cells, 20 (31%) in 1-49% of cells, and nine (14%) were PD-L1-negative. Non-synonymous TP53 mutations were observed in 41 (57%) and 25 (35%) harbored KRAS-mutated tumors. After a median follow-up of 15.2 months (95% confidence interval [CI] 10.3-17.4 m), the median OS in the TP53-mutated group was 18.1 months (95% CI 6.6-not reached), vs. 8.1 months (95% CI 2.2-14.5, hazard ratio [HR] = 0.48; 95% CI 0.25-0.95, p = 0.04) in the TP53-wild-type group. Median progression-free survival was significantly longer in TP53-mutated patients (4.5 months, 95% CI 2.8-18.1 versus 1.4, 95% CI 1.1-3.5; p = 0.03), although TP53 mutation status failed to significantly influence PFS in the multivariate analysis (p = 0.32). Objective response rate (ORR) was higher in patients with TP53 mutation (51.2% vs. 20.7%; p = 0.01). In multivariate analysis, TP53 mutations independently associated with longer OS (HR = 0.35, 95% CI 0.16-0.77, p = 0.009). CONCLUSIONS TP53-mutated status correlated with immunotherapy OS benefit in aNSCLC.
Collapse
Affiliation(s)
- Sandra Assoun
- Thoracic Oncology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France; U830 INSERM "Genetics and biology of cancers", Research Centre, Institut Curie, 26 rue d'Ulm, 75005, Paris, France
| | - Nathalie Theou-Anton
- Genetics Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France
| | - Marina Nguenang
- Thoracic Oncology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France
| | - Aurélie Cazes
- Pathology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France
| | - Claire Danel
- Pathology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France
| | - Baptiste Abbar
- Thoracic Oncology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France
| | - Johan Pluvy
- Thoracic Oncology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France
| | - Valérie Gounant
- Thoracic Oncology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France
| | - Antoine Khalil
- Radiology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France
| | - Céline Namour
- Thoracic Oncology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France
| | - Solenn Brosseau
- Thoracic Oncology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France; U830 INSERM "Genetics and biology of cancers", Research Centre, Institut Curie, 26 rue d'Ulm, 75005, Paris, France; Medicine Faculty, University Paris-Diderot, 46 rue Henri Huchard, 75018 Paris, France
| | - Gérard Zalcman
- Thoracic Oncology Department, University Hospital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris (AP-HP), 46 rue Henri Huchard, 75018, Paris, France; U830 INSERM "Genetics and biology of cancers", Research Centre, Institut Curie, 26 rue d'Ulm, 75005, Paris, France; Medicine Faculty, University Paris-Diderot, 46 rue Henri Huchard, 75018 Paris, France.
| |
Collapse
|
36
|
Zhang J, Tan P, Guo L, Gong J, Ma J, Li J, Lee M, Fang S, Jing J, Johnson G, Sun D, Cao WM, Dashwood R, Han L, Zhou Y, Dong WG, Huang Y. p53-dependent autophagic degradation of TET2 modulates cancer therapeutic resistance. Oncogene 2019; 38:1905-1919. [PMID: 30390073 PMCID: PMC6419514 DOI: 10.1038/s41388-018-0524-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 07/04/2018] [Accepted: 07/25/2018] [Indexed: 12/19/2022]
Abstract
Tumor cells with p53 inactivation frequently exhibit chemotherapy resistance, which poses a long-standing challenge to cancer treatment. Here we unveiled a previously unrecognized role of TET2 in mediating p53-loss induced chemotherapy resistance in colon cancer. Deletion of TET2 in p53-null colon cancer cells enhanced DNA damage and restored chemotherapy sensitivity. By taking a two-pronged approach that combined pharmacological inhibition with genetic depletion, we discovered that p53 destabilized TET2 at the protein level by promoting its autophagic degradation. At the molecular level, we further revealed a physical association between TET2 and p53 that facilitated the nucleoplasmic shuttling of TET2, as well as its recruitment to the autophagosome for degradation. Our study has unveiled a functional interplay between TET2 and p53 during anti-cancer therapy. Our findings establish the rationale for targeting TET2 to overcome chemotherapy resistance associated with mutant p53 tumors.
Collapse
Affiliation(s)
- Jixiang Zhang
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
- Department of gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Peng Tan
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Lei Guo
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Jing Gong
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jingjing Ma
- Department of gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Jia Li
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Minjung Lee
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Shaohai Fang
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Ji Jing
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Gavin Johnson
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Deqiang Sun
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Wen-Ming Cao
- Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China, 310022
| | - Roderick Dashwood
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
- Department of Molecular & Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Leng Han
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA.
- Department of Medical Physiology, College of Medicine, Texas A&M University, Temple, TX, 76504, USA.
| | - Wei-Guo Dong
- Department of gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China.
| | - Yun Huang
- Center for Epigenetics & Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA.
- Department of Molecular & Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
37
|
Gunaratna RT, Santos A, Luo L, Nagi C, Lambertz I, Spier M, Conti CJ, Fuchs-Young RS. Dynamic role of the codon 72 p53 single-nucleotide polymorphism in mammary tumorigenesis in a humanized mouse model. Oncogene 2019; 38:3535-3550. [PMID: 30651598 PMCID: PMC6756019 DOI: 10.1038/s41388-018-0630-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 09/14/2018] [Accepted: 11/23/2018] [Indexed: 12/16/2022]
Abstract
Female breast cancer (BrCa) is the most common noncutaneous cancer among women in the United States. Human epidemiological studies reveal that a p53 single-nucleotide polymorphism (SNP) at codon 72, encoding proline (P72) or arginine (R72), is associated with differential risk of several cancers, including BrCa. However, the molecular mechanisms by which these variants affect mammary tumorigenesis remain unresolved. To investigate the effects of this polymorphism on susceptibility to mammary cancer, we used a humanized p53 mouse model, homozygous for either P72 or R72. Our studies revealed that R72 mice had a significantly higher mammary tumor incidence and reduced latency in both DMBA-induced and MMTV-Erbb2/Neu mouse mammary tumor models compared to P72 mice. Analyses showed that susceptible mammary glands from E-R72 (R72 x MMTV-Erbb2/Neu) mice developed a senescence-associated secretory phenotype (SASP) with influx of proinflammatory macrophages, ultimately resulting in chronic, protumorigenic inflammation. Mammary tumors arising in E-R72 mice also had an increased influx of tumor-associated macrophages, contributing to angiogenesis and elevated tumor growth rates. These results demonstrate that the p53 R72 variant increased susceptibility to mammary tumorigenesis through chronic inflammation.
Collapse
Affiliation(s)
- Ramesh T Gunaratna
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX, USA.,Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, TX, USA.,Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Andres Santos
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, TX, USA.,Paul L. Foster School of Medicine, Texas Tech University Health Science Center, El Paso, TX, USA
| | - Linjie Luo
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, TX, USA
| | - Chandandeep Nagi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Isabel Lambertz
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, TX, USA
| | - Madison Spier
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, TX, USA
| | - Claudio J Conti
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, TX, USA.,Departamento de Bioingeniería, Universidad Carlos III de Madrid, Madrid, Spain.,Fundación Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain
| | - Robin S Fuchs-Young
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX, USA. .,Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, TX, USA.
| |
Collapse
|
38
|
Mantovani F, Collavin L, Del Sal G. Mutant p53 as a guardian of the cancer cell. Cell Death Differ 2019; 26:199-212. [PMID: 30538286 PMCID: PMC6329812 DOI: 10.1038/s41418-018-0246-9] [Citation(s) in RCA: 498] [Impact Index Per Article: 99.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/26/2018] [Accepted: 11/13/2018] [Indexed: 01/09/2023] Open
Abstract
Forty years of research have established that the p53 tumor suppressor provides a major barrier to neoplastic transformation and tumor progression by its unique ability to act as an extremely sensitive collector of stress inputs, and to coordinate a complex framework of diverse effector pathways and processes that protect cellular homeostasis and genome stability. Missense mutations in the TP53 gene are extremely widespread in human cancers and give rise to mutant p53 proteins that lose tumor suppressive activities, and some of which exert trans-dominant repression over the wild-type counterpart. Cancer cells acquire selective advantages by retaining mutant forms of the protein, which radically subvert the nature of the p53 pathway by promoting invasion, metastasis and chemoresistance. In this review, we consider available evidence suggesting that mutant p53 proteins can favor cancer cell survival and tumor progression by acting as homeostatic factors that sense and protect cancer cells from transformation-related stress stimuli, including DNA lesions, oxidative and proteotoxic stress, metabolic inbalance, interaction with the tumor microenvironment, and the immune system. These activities of mutant p53 may explain cancer cell addiction to this particular oncogene, and their study may disclose tumor vulnerabilities and synthetic lethalities that could be exploited for hitting tumors bearing missense TP53 mutations.
Collapse
Affiliation(s)
- Fiamma Mantovani
- Laboratorio Nazionale CIB (LNCIB), AREA Science Park, Trieste, Italy
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy
| | - Licio Collavin
- Laboratorio Nazionale CIB (LNCIB), AREA Science Park, Trieste, Italy
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy
| | - Giannino Del Sal
- Laboratorio Nazionale CIB (LNCIB), AREA Science Park, Trieste, Italy.
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste, Italy.
- IFOM-the FIRC Institute of Molecular Oncology, Trieste, Italy.
| |
Collapse
|
39
|
Regulators of Oncogenic Mutant TP53 Gain of Function. Cancers (Basel) 2018; 11:cancers11010004. [PMID: 30577483 PMCID: PMC6356290 DOI: 10.3390/cancers11010004] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/11/2018] [Accepted: 12/18/2018] [Indexed: 12/19/2022] Open
Abstract
The tumor suppressor p53 (TP53) is the most frequently mutated human gene. Mutations in TP53 not only disrupt its tumor suppressor function, but also endow oncogenic gain-of-function (GOF) activities in a manner independent of wild-type TP53 (wtp53). Mutant TP53 (mutp53) GOF is mainly mediated by its binding with other tumor suppressive or oncogenic proteins. Increasing evidence indicates that stabilization of mutp53 is crucial for its GOF activity. However, little is known about factors that alter mutp53 stability and its oncogenic GOF activities. In this review article, we primarily summarize key regulators of mutp53 stability/activities, including genotoxic stress, post-translational modifications, ubiquitin ligases, and molecular chaperones, as well as a single nucleotide polymorphism (SNP) and dimer-forming mutations in mutp53.
Collapse
|
40
|
Omrani-Nava V, Hedayatizadeh-Omran A, Alizadeh-Navaei R, Mokhberi V, Jalalian R, Janbabaei G, Amjadi O, Rahmatpour G, Mozaffari A. TP53 single nucleotide polymorphism (rs1042522) in Iranian patients with coronary artery disease. Biomed Rep 2018; 9:259-265. [PMID: 30271603 PMCID: PMC6158393 DOI: 10.3892/br.2018.1121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/22/2018] [Indexed: 02/05/2023] Open
Abstract
Chronic diseases including coronary artery disease (CAD) impose a high burden in terms of mortality and disability particularly in developing countries. Both genetic and environmental risk factors confer susceptibility to CAD. Meanwhile, a functional polymorphism in the tumor protein p53 (TP53) gene (codon 72, exon 4) has been reported to be associated with a wide range of cancers and inflammatory disorders. There are controversies regarding CAD and involvement of the TP53 codon 72 single nucleotide polymorphism; therefore, the present case-control study was conducted to evaluate the potential association between this TP53 polymorphism and CAD in an Iranian population. A total of 153 subjects (including 70 patients diagnosed with CAD and 83 subjects with normal coronary parameters, determined by angiography) were genotyped for the TP53 (rs1042522) polymorphism by the polymerase chain reaction-restriction fragment length polymorphism technique. Clinical and laboratory findings were also evaluated. The χ2 test and unpaired Student's t-test were applied to compare genotype and allele distributions and clinical characteristics between the two groups. Significant associations of the Pro72 allele [odds ratio (OR)=1.66, P=0.027] and Pro/Pro genotype (OR=2.91, P=0.022) with CAD were identified. No associations between patients' clinical findings and genotypes were apparent. Therefore, according to present findings, the TP53 Pro72 allele may be involved in the development of CAD along with conventional risk factors in patients from Northern Iran.
Collapse
Affiliation(s)
- Versa Omrani-Nava
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran 48166-33131, Iran
| | - Akbar Hedayatizadeh-Omran
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran 48166-33131, Iran
- Correspondence to: Dr Akbar Hedayatizadeh-Omran, Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, 70 Razi Street, Sari, Mazandaran 48166-33131, Iran, E-mail:
| | - Reza Alizadeh-Navaei
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran 48166-33131, Iran
| | - Vahid Mokhberi
- Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran 48471-91971, Iran
| | - Rozita Jalalian
- Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran 48471-91971, Iran
| | - Ghasem Janbabaei
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran 48166-33131, Iran
| | - Omolbanin Amjadi
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran 48166-33131, Iran
| | - Ghasem Rahmatpour
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran 48166-33131, Iran
| | - Amir Mozaffari
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Mazandaran 48166-33131, Iran
| |
Collapse
|
41
|
Lo Nigro C, Vivenza D, Denaro N, Lattanzio L, Fortunato M, Crook T, Merlano MC. DUSP2 methylation is a candidate biomarker of outcome in head and neck cancer. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:271. [PMID: 30094257 DOI: 10.21037/atm.2018.06.39] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Biomarkers predictive of response to chemoradiotherapy (CRT) regimens for locally advanced head and neck squamous cell carcinoma (LA-HNSCC) are urgently required to identify patients in whom this approach is likely to be effective. TP53 mutations and epidermal growth factor (EGFR) overexpression are common markers of disease. Dual-specificity-phosphatase-2 (DUSP2) has an essential role in cell proliferation, cancer and immune responses. Methods Aberrant DUSP2 methylation was investigated by pyrosequencing in 5 HNSCC cell lines, 112 LA-HNSCC tumours. EGFR was investigated by immunohistochemistry and TP53 was analysed by sequencing. Results We demonstrate methylation-dependent transcriptional silencing of DUSP2 in HNSCC cell lines. In LA-HNSCC patients, aberrant methylation in the DUSP2 CpG island was present in 51/112 cases (45.5%). LA-HNSCC cases with wild-type TP53, overexpression of EGFR and unmethylated DUSP2 had the worst overall survival (P≤0.001). Conclusions DUSP2 methylation, when combined with EGFR and TP53, is a candidate biomarker of clinical outcome in LA-HNSCC treated with CRT.
Collapse
Affiliation(s)
- Cristiana Lo Nigro
- Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Daniela Vivenza
- Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Nerina Denaro
- Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Laura Lattanzio
- Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Mirella Fortunato
- Department of Pathology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| | - Tim Crook
- Department of Oncology, St. Lukes Cancer Centre, Guildford, UK
| | - Marco Carlo Merlano
- Medical Oncology, Department of Oncology, S. Croce & Carle Teaching Hospital, Cuneo, Italy
| |
Collapse
|
42
|
TP53 rs1625895 is Related to Breast Cancer Incidence and Early Death in Iranian Population. Indian J Clin Biochem 2018; 34:485-489. [PMID: 31686737 DOI: 10.1007/s12291-018-0774-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/16/2018] [Indexed: 12/29/2022]
Abstract
Breast cancer as the second most common cancer worldwide tend to be experienced by Iranian women 10 years earlier with a peak incidence at the premenopausal stage. Genetic mutations of TP53 tumor suppressor gene has been shown to be related to early onset of breast cancer. It has been shown already that rs1625895 polymorphic site is related to glioma as well as lung cancer. In this study, we have investigated the role of rs1625895 polymorphism in breast cancer incidence in Iranian women. DNA extraction of 86 breast cancer patients and 96 control individuals have been used for allele-specific primer-PCR and genotyping of allele A and allele G of the TP53 rs1625895. Genotypes frequencies have been shown that GG homozygosis as the most frequent genotype is a significant association with increased risk of breast cancer development in Iranian women (odds ratio = 6, p = 0.002). On the other hand and in comparison to allele G, allele A could cause early death of breast cancer patients by threefolds significantly (p = 0.011). As a conclusion, we show that allele A is the minor allele in both breast cancer patients and also control individuals and major allele G, is related to the increased risk of breast cancer development in Iranian women.
Collapse
|
43
|
Bellazzo A, Sicari D, Valentino E, Del Sal G, Collavin L. Complexes formed by mutant p53 and their roles in breast cancer. BREAST CANCER-TARGETS AND THERAPY 2018; 10:101-112. [PMID: 29950894 PMCID: PMC6011883 DOI: 10.2147/bctt.s145826] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Breast cancer is the most frequently diagnosed malignancy in women, and mutations in the tumor suppressor p53 are commonly detected in the most aggressive subtypes. The majority of TP53 gene alterations are missense substitutions, leading to expression of mutant forms of the p53 protein that are frequently detected at high levels in cancer cells. P53 mutants not only lose the physiological tumor-suppressive activity of the wild-type p53 protein but also acquire novel powerful oncogenic functions, referred to as gain of function, that may actively confer a selective advantage during tumor progression. Some of the best-characterized oncogenic activities of mutant p53 are mediated by its ability to form aberrant protein complexes with other transcription factors or proteins not directly related to gene transcription. The set of cellular proteins available to interact with mutant p53 is dependent on cell type and extensively affected by environmental signals, so the prognostic impact of p53 mutation is complex. Specific functional interactions of mutant p53 can profoundly impact homeostasis of breast cancer cells, reprogramming gene expression in response to specific extracellular inputs or cell-intrinsic conditions. The list of protein complexes involving mutant p53 in breast cancer is continuously growing, as is the number of oncogenic phenotypes in which they could be involved. In consideration of the functional impact of such complexes, key interactions of mutant p53 may be exploited as potential targets for development of therapies aimed at defusing the oncogenic potential of p53 mutation.
Collapse
Affiliation(s)
- Arianna Bellazzo
- National Laboratory CIB (LNCIB), AREA Science park, Trieste, Italy
| | - Daria Sicari
- National Laboratory CIB (LNCIB), AREA Science park, Trieste, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Elena Valentino
- National Laboratory CIB (LNCIB), AREA Science park, Trieste, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Giannino Del Sal
- National Laboratory CIB (LNCIB), AREA Science park, Trieste, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Licio Collavin
- National Laboratory CIB (LNCIB), AREA Science park, Trieste, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
44
|
Coelho A, Nogueira A, Soares S, Assis J, Pereira D, Bravo I, Catarino R, Medeiros R. TP53 Arg72Pro polymorphism is associated with increased overall survival but not response to therapy in Portuguese/Caucasian patients with advanced cervical cancer. Oncol Lett 2018; 15:8165-8171. [PMID: 29731921 DOI: 10.3892/ol.2018.8354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 02/09/2018] [Indexed: 11/05/2022] Open
Abstract
Identification of mechanisms that influence the therapeutic response and survival in patients with cancer is important. It is known that the genetic variability of the host, including presence of genetic polymorphisms in genes involved in DNA damage response, serves a crucial role in the prognosis of these patients. The present hospital-based retrospective cohort study aimed to evaluate the influence of TP53 Arg72Pro (rs1042522) polymorphism in the clinical outcome of 260 Caucasian patients diagnosed with cervical cancer and treated with concomitant radiotherapy and chemotherapy. The polymorphism genotyping was assessed using allelic discrimination by quantiative polymerase chain reaction. The results indicate that the TP53 Arg72Pro polymorphism did not significantly impact the response to therapy (P=0.571) nor disease-free survival (P=0.081). However, the polymorphism did influence overall survival, as increased median survival time was observed for patients carrying Arg/Pro genotype when compared with patients with Arg/Arg and Pro/Pro genotypes (126 months vs. 111 months, respectively; P=0.047). To conclude, the present findings suggest that a pharmacogenomic profile based on the genetic background of patients, including the analysis of the TP53 genotypes, may individualize treatment nad assist in the selection of therapies that may improve clinical outcome and lower toxicity for the patients.
Collapse
Affiliation(s)
- Ana Coelho
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Augusto Nogueira
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Sílvia Soares
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Joana Assis
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Deolinda Pereira
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,Oncology Department, Portuguese Oncology Institute of Porto, 4200-072 Porto, Portugal
| | - Isabel Bravo
- Medical Physics, Radiobiology and Radioprotection Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, 4200-072 Porto, Portugal
| | - Raquel Catarino
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto-Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.,CEBIMED, Faculty of Health Sciences of Fernando Pessoa University, 4249-004 Porto, Portugal.,Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
45
|
Mascelli S, Nozza P, Jones DTW, Colin C, Pistorio A, Milanaccio C, Ravegnani M, Consales A, Witt O, Morana G, Cama A, Capra V, Biassoni R, Pfister SM, Figarella-Branger D, Garrè ML, Raso A. TP53 codon 72 polymorphism may predict early tumour progression in paediatric pilocytic astrocytoma. Oncotarget 2018; 7:47918-47926. [PMID: 27374106 PMCID: PMC5216988 DOI: 10.18632/oncotarget.10295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 05/10/2016] [Indexed: 12/13/2022] Open
Abstract
Pilocytic astrocytoma and ganglioglioma may occur in inaccessible or surgically difficult areas. In case of incomplete resection, the availability of biological predictors of tumour progression could be particularly important. To this end, an analysis of p53 codon 72 polymorphism and assessment of its role as prognostic marker were performed. The status of the p53 Arg72Pro polymorphism was evaluated by pyrosequencing method in a multicenter cohort of 170 paediatric patients. Genotype/phenotype associations were investigated either by means of bivariate or multivariate analyses. In the partially resected pilocytic astrocytomas, the Arg/Arg variant predicts early tumour progression (median survival time: 23.1 months) and is associated with poor event-free survival (p value = 0.0009). This finding remains true also in case of adjuvant therapies, with a 5-year event-free survival of 30.6% for cases with Arg/Arg variant vs. 78.7% for those with other genotypes. There is no association between ganglioglioma and the polymorphism. The assessment of Arg/Arg variant could improve the management of pilocytic astrocytoma. TP53 codon 72 analysis could distinguish low-risk cases, in which surgery could be conservative, from high-risk cases needing an aggressive surgery plan.
Collapse
Affiliation(s)
- Samantha Mascelli
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Paolo Nozza
- UOC Anatomia Patologica, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - David T W Jones
- Division of Paediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Carole Colin
- CRO2 UMR_S911, Inserm, Aix-Marseille Université, 13385 Marseille, France
| | - Angela Pistorio
- Epidemiologia, Biostatistica e Comitati, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Claudia Milanaccio
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Marcello Ravegnani
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Alessandro Consales
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Olaf Witt
- Department of Paediatric Oncology, Haematology and Immunology, University of Heidelberg, Heidelberg, 69120 Heidelberg, Germany
| | - Giovanni Morana
- Neuroradiologia Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Armando Cama
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Valeria Capra
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Roberto Biassoni
- Laboratory Molecular Medicine, Translational Medicine Department, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Stefan M Pfister
- Division of Paediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany.,Department of Paediatric Oncology, Haematology and Immunology, University of Heidelberg, Heidelberg, 69120 Heidelberg, Germany
| | - Dominique Figarella-Branger
- CRO2 UMR_S911, Inserm, Aix-Marseille Université, 13385 Marseille, France.,APHM, Hôpital de la Timone, Service d'Anatomie Pathologique et de Neuropathologie, 13385 Marseille, France
| | - Maria Luisa Garrè
- Centro di Neuro-Oncologia, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Alessandro Raso
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| |
Collapse
|
46
|
The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget 2018; 8:8921-8946. [PMID: 27888811 PMCID: PMC5352454 DOI: 10.18632/oncotarget.13475] [Citation(s) in RCA: 379] [Impact Index Per Article: 63.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 10/13/2016] [Indexed: 01/10/2023] Open
Abstract
Cancer has long been a grievous disease complicated by innumerable players aggravating its cure. Many clinical studies demonstrated the prognostic relevance of the tumor suppressor protein p53 for many human tumor types. Overexpression of mutated p53 with reduced or abolished function is often connected to resistance to standard medications, including cisplatin, alkylating agents (temozolomide), anthracyclines, (doxorubicin), antimetabolites (gemcitabine), antiestrogenes (tamoxifen) and EGFR-inhibitors (cetuximab). Such mutations in the TP53 gene are often accompanied by changes in the conformation of the p53 protein. Small molecules that restore the wild-type conformation of p53 and, consequently, rebuild its proper function have been identified. These promising agents include PRIMA-1, MIRA-1, and several derivatives of the thiosemicarbazone family. In addition to mutations in p53 itself, p53 activity may be also be impaired due to alterations in p53s regulating proteins such as MDM2. MDM2 functions as primary cellular p53 inhibitor and deregulation of the MDM2/p53-balance has serious consequences. MDM2 alterations often result in its overexpression and therefore promote inhibition of p53 activity. To deal with this problem, a judicious approach is to employ MDM2 inhibitors. Several promising MDM2 inhibitors have been described such as nutlins, benzodiazepinediones or spiro-oxindoles as well as novel compound classes such as xanthone derivatives and trisubstituted aminothiophenes. Furthermore, even naturally derived inhibitor compounds such as a-mangostin, gambogic acid and siladenoserinols have been discovered. In this review, we discuss in detail such small molecules that play a pertinent role in affecting the p53-MDM2 signaling axis and analyze their potential as cancer chemotherapeutics.
Collapse
|
47
|
Gingerich MA, Smith JD, Michmerhuizen NL, Ludwig M, Devenport S, Matovina C, Brenner C, Chinn SB. Comprehensive review of genetic factors contributing to head and neck squamous cell carcinoma development in low-risk, nontraditional patients. Head Neck 2018; 40:943-954. [PMID: 29427520 DOI: 10.1002/hed.25057] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/22/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The past 2 decades have seen an increased incidence of head and neck squamous cell carcinoma (HNSCC) in a nontraditional, low-risk patient population (ie, ≤45 years of age, no substance use history), owing to a combination of human papillomavirus (HPV) infection and individual genetic variation. METHODS Articles positing genetic variants as contributing factors in HNSCC incidence in low-risk, nontraditional patients were identified using a PubMed search, reviewed in detail, and concisely summarized herein. RESULTS Recent data suggest that common polymorphisms in DNA repair enzymes, cell-cycle control proteins, apoptotic pathway members, and Fanconi anemia-associated genes likely modulate susceptibility to HNSCC development in low-risk, nontraditional patients. CONCLUSION At present, there is a lack of robust, comprehensive data on genetic drivers of oncogenesis in low-risk patients and a clear need for further research on genetic alterations underlying the rising incidence of HNSCC in low-risk, nontraditional patients.
Collapse
Affiliation(s)
- Morgan A Gingerich
- Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Joshua D Smith
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Nicole L Michmerhuizen
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Megan Ludwig
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan.,Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Samantha Devenport
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan.,Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Chloe Matovina
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Chad Brenner
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan.,Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| | - Steven B Chinn
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan.,Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
48
|
Uno M, Oba-Shinjo SM, Wakamatsu A, Huang N, Ferreira Alves VA, Rosemberg S, de Aguiar P, Leite C, Miura F, Marino RJ, Scaff M, Nagahashi-Marie SK. Association of TP53 Mutation, p53 Overexpression, and p53 Codon 72 Polymorphism with Susceptibility to Apoptosis in Adult Patients with Diffuse Astrocytomas. Int J Biol Markers 2018; 21:50-7. [PMID: 16711514 DOI: 10.1177/172460080602100108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Clarification of TP53 alterations is important to understand the mechanisms underlying the development of diffuse astrocytomas. It has been suggested that the alleles of TP53 at codon 72 differ in their ability to induce apoptosis in human cancers. The aim of this study was to analyze the possible association of TP53 mutation, p53 overexpression, and p53 codon 72 polymorphism with susceptibility to apoptosis in adult Brazilian patients with diffuse astrocytomas. We analyzed 56 surgical specimens of diffuse astrocytomas for alterations of TP53, using polymerase chain reaction single-strand conformation polymorphism (PCR-SSCP) direct sequencing. p53 and cleaved caspase 3 protein expression were assessed by immunohistochemistry. We found TP53 mutations in 19.6% (11 out of 56) of tumors tested, with the lowest mutation rate found in the cases of glioblastomas (8.8%) (p = 0.03). Only 16.1% of tumors tested showed cleaved caspase 3-positive staining, demonstrating that apoptosis is very inhibited in these tumors. All tumors having TP53 mutation and p53 accumulation had no expression of cleaved caspase 3. Additionally, no association was observed in tumors having proline and arginine alleles and expression of cleaved caspase 3. We concluded that clarification of the TP53 alterations allows a better understanding of the mechanisms involved in the progression of diffuse astrocytomas, and the allele status at codon 72 was not associated with apoptosis in these tumors.
Collapse
Affiliation(s)
- M Uno
- Department of Neurology, School of Medicine, University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ruwali M. Role of Genetic Variations in Determining Treatment Outcome in Head and Neck Cancer. EUROPEAN MEDICAL JOURNAL 2017. [DOI: 10.33590/emj/10313625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Worldwide, head and neck squamous cell carcinoma (HNSCC) is responsible for >550,000 diagnoses and 380,000 deaths annually. It originates in the upper aerodigestive tract and has a multifactorial origin involving both genetic and lifestyle risk factors. The clinical management of HNSCC involves surgery, radiotherapy, and chemotherapy. Several studies point to the role of genetic variations in predicting drug efficacy and toxicity. Cancer pharmacogenomics has fast emerged as a new and promising field for the early identification of genetic markers that can predict drug response or toxicity, with the number of studies of genetic polymorphisms as prognostic factors of HNSCC treatment outcomes growing. The number of studies evaluating the association of candidate polymorphisms in drug-metabolising Phase I and II enzymes with treatment outcome far exceed the studies involving other candidate genes, such as those involved in drug metabolism, DNA repair, and cell cycle regulation. This review focusses on the relevance of genetic variations in genes, where the corresponding gene products play an important role in drug metabolism (TPMT, DPD), DNA repair (X-ray repair cross complementing 1), cell cycle (tumour protein P53), and carcinogenesis (matrix metalloproteinase 3 and 7), thereby contributing to the treatment outcome for HNSCC. This could greatly help clinicians in identifying genetic markers useful for the selection of optimal drugs, dose, and treatment duration on an individual basis, resulting in improved drug efficacy and decreased toxicity. However, further studies are needed in well characterised and larger HNSCC populations with proper validation of pharmacogenetic markers in experimental settings before application in clinical routine diagnostics.
Collapse
Affiliation(s)
- Munindra Ruwali
- Amity Institute of Biotechnology, Amity University Haryana, Gurgaon (Manesar), India
| |
Collapse
|
50
|
Saensa-ard S, Leuangwattanawanit S, Senggunprai L, Namwat N, Kongpetch S, Chamgramol Y, Loilome W, Khansaard W, Jusakul A, Prawan A, Pairojkul C, Khantikeo N, Yongvanit P, Kukongviriyapan V. Establishment of cholangiocarcinoma cell lines from patients in the endemic area of liver fluke infection in Thailand. Tumour Biol 2017; 39:1010428317725925. [DOI: 10.1177/1010428317725925] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinoma is a rare type of cancer which is an increasingly discernible health threat. The disease is usually very difficult in diagnosis and various treatment modalities are typically not effective. Cholangiocarcinoma is a complex and very heterogeneous malignancy characterized by tumor location, different risk factors, molecular profiling, and prognosis. Cancer cell lines represent an important tool for investigation in various aspects of tumor biology and molecular therapeutics. We established two cell lines, KKU-452 and KKU-023, which were derived from patients residing in the endemic area of liver fluke infection in Thailand. Both of tumor tissues have gross pathology of perihilar and intrahepatic mass-forming cholangiocarcinoma. Two cell lines were characterized for their biological, molecular and genetic properties. KKU-452 and KKU-023 cells are both adherent cells with epithelium morphology, but have some differences in their growth pattern (a doubling time of 17.9 vs 34.8 h, respectively) and the expression of epithelial bile duct markers, CK7 and CK19. Cytogenetic analysis of KKU-452 and KKU-023 cells revealed their highly complex karyotypes; hypertriploid and hypotetraploid, respectively, with multiple chromosomal aberrations. Both cell lines showed mutations in p53 but not in KRAS. KKU-452 showed a very rapid migration and invasion properties in concert with low expression of E-cadherin and high expression of N-cadherin, whereas KKU-023 showed opposite characters. KKU-023, but not KKU-452, showed in vivo tumorigenicity in xenografted nude mice. Those two established cholangiocarcinoma cell lines with unique characters may be valuable for better understanding the process of carcinogenesis and developing new therapeutics for the patients
Collapse
Affiliation(s)
- Sunitta Saensa-ard
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | | | - Laddawan Senggunprai
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nisana Namwat
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sarinya Kongpetch
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Yaovalux Chamgramol
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharin Loilome
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Walaiporn Khansaard
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Apinya Jusakul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Auemduan Prawan
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chawalit Pairojkul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Narong Khantikeo
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Puangrat Yongvanit
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Veerapol Kukongviriyapan
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|