1
|
Picard M, Scott-Boyer MP, Bodein A, Leclercq M, Prunier J, Périn O, Droit A. Target repositioning using multi-layer networks and machine learning: The case of prostate cancer. Comput Struct Biotechnol J 2024; 24:464-475. [PMID: 38983753 PMCID: PMC11231507 DOI: 10.1016/j.csbj.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024] Open
Abstract
The discovery of novel therapeutic targets, defined as proteins which drugs can interact with to induce therapeutic benefits, typically represent the first and most important step of drug discovery. One solution for target discovery is target repositioning, a strategy which relies on the repurposing of known targets for new diseases, leading to new treatments, less side effects and potential drug synergies. Biological networks have emerged as powerful tools for integrating heterogeneous data and facilitating the prediction of biological or therapeutic properties. Consequently, they are widely employed to predict new therapeutic targets by characterizing potential candidates, often based on their interactions within a Protein-Protein Interaction (PPI) network, and their proximity to genes associated with the disease. However, over-reliance on PPI networks and the assumption that potential targets are necessarily near known genes can introduce biases that may limit the effectiveness of these methods. This study addresses these limitations in two ways. First, by exploiting a multi-layer network which incorporates additional information such as gene regulation, metabolite interactions, metabolic pathways, and several disease signatures such as Differentially Expressed Genes, mutated genes, Copy Number Alteration, and structural variants. Second, by extracting relevant features from the network using several approaches including proximity to disease-associated genes, but also unbiased approaches such as propagation-based methods, topological metrics, and module detection algorithms. Using prostate cancer as a case study, the best features were identified and utilized to train machine learning algorithms to predict 5 novel promising therapeutic targets for prostate cancer: IGF2R, C5AR, RAB7, SETD2 and NPBWR1.
Collapse
Affiliation(s)
- Milan Picard
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Marie-Pier Scott-Boyer
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Antoine Bodein
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Mickaël Leclercq
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Julien Prunier
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Olivier Périn
- Digital Transformation and Innovation Department, L'Oréal Advanced Research, Aulnay-sous-bois, France
| | - Arnaud Droit
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| |
Collapse
|
2
|
VanSlyke JK, Boswell BA, Musil LS. ErbBs in Lens Cell Fibrosis and Secondary Cataract. Invest Ophthalmol Vis Sci 2023; 64:6. [PMID: 37418274 PMCID: PMC10337807 DOI: 10.1167/iovs.64.10.6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/30/2023] [Indexed: 07/08/2023] Open
Abstract
Purpose TGFβ-induced epithelial-to-myofibroblast transition (EMyT) of lens cells has been linked to the most common vision-disrupting complication of cataract surgery-namely, posterior capsule opacification (PCO; secondary cataract). Although inhibitors of the ErbB family of receptor tyrosine kinases have been shown to block some PCO-associated processes in model systems, our knowledge of ErbB signaling in the lens is very limited. Here, we investigate the expression of ErbBs and their ligands in primary cultures of chick lens epithelial cells (dissociated cell-derived monolayer cultures [DCDMLs]) and how TGFβ affects ErbB function. Methods DCDMLs were analyzed by immunofluorescence microscopy and Western blotting under basal and profibrotic conditions. Results Small-molecule ErbB kinase blockers, including the human therapeutic lapatinib, selectively inhibit TGFβ-induced EMyT of DCDMLs. Lens cells constitutively express ErbB1 (EGFR), ErbB2, and ErbB4 protein on the plasma membrane and release into the medium ErbB-activating ligand. Culturing DCDMLs with TGFβ increases soluble bioactive ErbB ligand and markedly alters ErbBs, reducing total and cell surface ErbB2 and ErbB4 while increasing ErbB1 expression and homodimer formation. Similar, TGFβ-dependent changes in relative ErbB expression are induced when lens cells are exposed to the profibrotic substrate fibronectin. A single, 1-hour treatment with lapatinib inhibits EMyT in DCDMLs assessed 6 days later. Short-term exposure to lower doses of lapatinib is also capable of eliciting a durable response when combined with suboptimal levels of a mechanistically distinct multikinase inhibitor. Conclusions Our findings support ErbB1 as a therapeutic target for fibrotic PCO, which could be leveraged to pharmaceutically preserve the vision of millions of patients with cataracts.
Collapse
Affiliation(s)
- Judy K. VanSlyke
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Bruce A. Boswell
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Linda S. Musil
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
3
|
Barés G, Beà A, Hernández L, Navaridas R, Felip I, Megino C, Blasco N, Nadeu F, Campo E, Llovera M, Dolcet X, Sanchis D. ENDOG Impacts on Tumor Cell Proliferation and Tumor Prognosis in the Context of PI3K/PTEN Pathway Status. Cancers (Basel) 2021; 13:3803. [PMID: 34359707 PMCID: PMC8345062 DOI: 10.3390/cancers13153803] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 11/17/2022] Open
Abstract
EndoG influences mitochondrial DNA replication and is involved in somatic cell proliferation. Here, we investigated the effect of ENDOG/Endog expression on proliferation in different tumor models. Noteworthy, ENDOG deficiency reduced proliferation of endometrial tumor cells expressing low PTEN/high p-AKT levels, and Endog deletion blunted the growth of PTEN-deficient 3D endometrial cultures. Furthermore, ENDOG silencing reduced proliferation of follicular thyroid carcinoma and glioblastoma cell lines with high p-AKT expression. High ENDOG expression was associated with a short time to treatment in a cohort of patients with chronic lymphocytic leukemia (CLL), a B-cell lymphoid neoplasm with activation of PI3K/AKT. This clinical impact was observed in the less aggressive CLL subtype with mutated IGHV in which high ENDOG and low PTEN levels were associated with worse outcome. In summary, our results show that reducing ENDOG expression hinders growth of some tumors characterized by low PTEN activity and high p-AKT expression and that ENDOG has prognostic value for some cancer types.
Collapse
Affiliation(s)
- Gisel Barés
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida-IRBLleida, 25198 Lleida, Spain; (G.B.); (A.B.); (N.B.); (M.L.)
| | - Aida Beà
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida-IRBLleida, 25198 Lleida, Spain; (G.B.); (A.B.); (N.B.); (M.L.)
| | - Luís Hernández
- Lymphoid Neoplasm Program, Institut d’Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS) and CIBERONC, 08036 Barcelona, Spain; (F.N.); (E.C.)
| | - Raul Navaridas
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida–IRBLleida and CIBERONC, 25198 Lleida, Spain; (R.N.); (I.F.); (C.M.); (X.D.)
| | - Isidre Felip
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida–IRBLleida and CIBERONC, 25198 Lleida, Spain; (R.N.); (I.F.); (C.M.); (X.D.)
| | - Cristina Megino
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida–IRBLleida and CIBERONC, 25198 Lleida, Spain; (R.N.); (I.F.); (C.M.); (X.D.)
| | - Natividad Blasco
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida-IRBLleida, 25198 Lleida, Spain; (G.B.); (A.B.); (N.B.); (M.L.)
| | - Ferran Nadeu
- Lymphoid Neoplasm Program, Institut d’Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS) and CIBERONC, 08036 Barcelona, Spain; (F.N.); (E.C.)
| | - Elías Campo
- Lymphoid Neoplasm Program, Institut d’Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS) and CIBERONC, 08036 Barcelona, Spain; (F.N.); (E.C.)
- Department of Oncology, Hospital Clinic of Barcelona, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Marta Llovera
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida-IRBLleida, 25198 Lleida, Spain; (G.B.); (A.B.); (N.B.); (M.L.)
| | - Xavier Dolcet
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida–IRBLleida and CIBERONC, 25198 Lleida, Spain; (R.N.); (I.F.); (C.M.); (X.D.)
| | - Daniel Sanchis
- Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida-IRBLleida, 25198 Lleida, Spain; (G.B.); (A.B.); (N.B.); (M.L.)
| |
Collapse
|
4
|
Casadei-Gardini A, Vagheggini A, Gelsomino F, Spallanzani A, Ulivi P, Orsi G, Rovesti G, Andrikou K, Tamburini E, Scartozzi M, Cascinu S. Is There an Optimal Choice in Refractory Colorectal Cancer? A Network Meta-Analysis. Clin Colorectal Cancer 2020; 19:82-90.e9. [PMID: 32192883 DOI: 10.1016/j.clcc.2019.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND In the absence of head-to-head comparison studies, the present network meta-analysis evaluated and compared the efficacy of 4 therapeutic alternatives for refractory colorectal cancer. MATERIALS AND METHODS The search focused on results from phase III randomized controlled trials. Separate (subgroup) network meta-analyses were conducted to obtain drug comparisons stratified by various patient characteristics. The principal outcome of interest was overall survival (OS). RESULTS No difference in OS was found between regorafenib and TAS-102. For a rectal primary location, TAS-102 conferred benefit versus placebo (hazard ratio [HR], 0.671), but regorafenib did not (HR, 0.950). For patients aged > 65 years, TAS-102 showed benefit versus placebo (HR, 0.579) but regorafenib did not (HR, 0.816). For patients with an Eastern Cooperative Oncology Group (ECOG) performance status (PS) of 0 in the indirect comparison, regorafenib showed benefit versus placebo (HR, 0.687), as did TAS-102 (HR, 0.756) but with a lower advantage. For patients with RAS wild type not previously treated with anti-EGFR antibodies, panitumumab was the optimal choice for OS. CONCLUSIONS No differences in OS were found between regorafenib and TAS-102. Possible greater efficacy was found for TAS-102 compared with regorafenib for patients with a rectal primary location, ECOG PS > 0, and age > 65 years. In contrast, regorafenib showed possible greater effectiveness for patients with ECOG PS 0 and age < 65 years. In the RAS WT population, the anti-EGFR drug showed superiority with respect to TAS-102 and regorafenib. These results should be viewed as only exploratory, and further prospective studies are warranted to validate these data.
Collapse
Affiliation(s)
- Andrea Casadei-Gardini
- Division of Oncology, Department of Oncology and Hematology, University Hospital Modena, Modena, Italy.
| | - Alessandro Vagheggini
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, IRCCS, Meldola, Italy
| | - Fabio Gelsomino
- Division of Oncology, Department of Oncology and Hematology, University Hospital Modena, Modena, Italy
| | - Andrea Spallanzani
- Division of Oncology, Department of Oncology and Hematology, University Hospital Modena, Modena, Italy
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, IRCCS, Meldola, Italy
| | - Giulia Orsi
- Division of Oncology, Department of Oncology and Hematology, University Hospital Modena, Modena, Italy
| | - Giulia Rovesti
- Division of Oncology, Department of Oncology and Hematology, University Hospital Modena, Modena, Italy
| | - Kalliopi Andrikou
- Division of Oncology, Department of Oncology and Hematology, University Hospital Modena, Modena, Italy
| | - Emiliano Tamburini
- Department of Medical Oncology, Cardinale Giovanni Panico Hospital of Tricase, Tricase, Italy
| | - Mario Scartozzi
- Department of Medical Oncology, "Duilio Casula" Polyclinic, Cagliari State University, Cagliari, Italy
| | - Stefano Cascinu
- Division of Oncology, Department of Oncology and Hematology, University Hospital Modena, Modena, Italy
| |
Collapse
|
5
|
Jin Y, Liu M, Sa R, Fu H, Cheng L, Chen L. Mouse models of thyroid cancer: Bridging pathogenesis and novel therapeutics. Cancer Lett 2019; 469:35-53. [PMID: 31589905 DOI: 10.1016/j.canlet.2019.09.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/25/2019] [Accepted: 09/30/2019] [Indexed: 12/27/2022]
Abstract
Due to a global increase in the incidence of thyroid cancer, numerous novel mouse models were established to reveal thyroid cancer pathogenesis and test promising therapeutic strategies, necessitating a comprehensive review of translational medicine that covers (i) the role of mouse models in the research of thyroid cancer pathogenesis, and (ii) preclinical testing of potential anti-thyroid cancer therapeutics. The present review article aims to: (i) describe the current approaches for mouse modeling of thyroid cancer, (ii) provide insight into the biology and genetics of thyroid cancers, and (iii) offer guidance on the use of mouse models for testing potential therapeutics in preclinical settings. Based on research with mouse models of thyroid cancer pathogenesis involving the RTK, RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, SRC, and JAK-STAT signaling pathways, inhibitors of VEGFR, MEK, mTOR, SRC, and STAT3 have been developed as anti-thyroid cancer drugs for "bench-to-bedside" translation. In the future, mouse models of thyroid cancer will be designed to be ''humanized" and "patient-like," offering opportunities to: (i) investigate the pathogenesis of thyroid cancer through target screening based on the CRISPR/Cas system, (ii) test drugs based on new mouse models, and (iii) explore the underlying mechanisms based on multi-omics.
Collapse
Affiliation(s)
- Yuchen Jin
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| | - Min Liu
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China; Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai, 200032, China.
| | - Ri Sa
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| | - Hao Fu
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| | - Lin Cheng
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| | - Libo Chen
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| |
Collapse
|
6
|
Fondevila F, Méndez-Blanco C, Fernández-Palanca P, González-Gallego J, Mauriz JL. Anti-tumoral activity of single and combined regorafenib treatments in preclinical models of liver and gastrointestinal cancers. Exp Mol Med 2019; 51:1-15. [PMID: 31551425 PMCID: PMC6802659 DOI: 10.1038/s12276-019-0308-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/09/2019] [Accepted: 06/12/2019] [Indexed: 12/24/2022] Open
Abstract
Regorafenib is a sorafenib-derived chemotherapy drug belonging to the multikinase inhibitor family. This agent effectively targets a wide range of tyrosine kinases involved in cancer biology, such as those implicated in oncogenesis, angiogenesis, and tumor microenvironment control. The beneficial effects of regorafenib in clinical trials of patients who suffer from advanced hepatocellular carcinoma (HCC), colorectal cancer (CRC) or gastrointestinal stromal tumors (GISTs) refractory to standard treatments led to regorafenib monotherapy approval as a second-line treatment for advanced HCC and as a third-line treatment for advanced CRC and GISTs. Multiple in vitro and in vivo studies have been performed over the last decade to reveal the molecular mechanisms of the favorable actions exerted by regorafenib in patients. Given the hypothetical loss of sensitivity to regorafenib in tumor cells, preclinical research is also searching for novel therapeutic approaches consisting of co-administration of this drug plus other agents as a strategy to improve regorafenib effectiveness. This review summarizes the anti-tumor effects of regorafenib in single or combined treatment in preclinical models of HCC, CRC and GISTs and discusses both the global and molecular effects that account for its anti-cancer properties in the clinical setting. The cancer drug regorafenib exhibits a broad range of anti-tumor activities that could be enhanced by combination with other treatments. A team led by José L. Mauriz from the University of León, Spain, review the ways in which regorafenib, blocking several enzymes involved in cancer biology, has been shown to shrink tumors in different models of liver, colon and gastrointestinal cancer. Its mechanisms of action include blockade of new blood vessel formation, induction of cell death and modulation of the immune microenvironment. Research studies show that co-administration of regorafenib with other drugs directed at various molecular targets or immune pathways produces synergistic effects against cancer cells. The preclinical data highlights the potential of combination drug regimens to improve outcomes among patients eligible for regorafenib treatment.
Collapse
Affiliation(s)
- Flavia Fondevila
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Carolina Méndez-Blanco
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Paula Fernández-Palanca
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Javier González-Gallego
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - José L Mauriz
- Institute of Biomedicine, University of León, León, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain.
| |
Collapse
|
7
|
Huot JR, Essex AL, Gutierrez M, Barreto R, Wang M, Waning DL, Plotkin LI, Bonetto A. Chronic Treatment with Multi-Kinase Inhibitors Causes Differential Toxicities on Skeletal and Cardiac Muscles. Cancers (Basel) 2019; 11:cancers11040571. [PMID: 31018508 PMCID: PMC6520777 DOI: 10.3390/cancers11040571] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/12/2019] [Accepted: 04/18/2019] [Indexed: 12/20/2022] Open
Abstract
Despite recent progress, chemotherapy remains the preferred treatment for cancer. We have shown a link between anticancer drugs and the development of cachexia, i.e., body wasting accompanied by muscle loss. The multi-kinase inhibitors (MKIs) regorafenib and sorafenib, used as second-line treatment for solid tumors, are frequently accompanied by several side effects, including loss of muscle mass and strength. In the present study we aimed to investigate the molecular mechanisms associated with the occurrence of muscle toxicities in in vivo conditions. Hence, we treated 8-week old healthy CD2F1 male mice with MKIs for up to six weeks and observed decreased skeletal and cardiac muscle mass, consistent with muscle weakness. Modulation of ERK1/2 and GSK3β, as well as increased expression of markers of autophagy, previously associated with muscle atrophy conditions, were shown in skeletal muscle upon treatment with either drug. MKIs also promoted cardiac abnormalities consistent with reduced left ventricular mass, internal diameter, posterior wall thickness and stroke volume, despite unchanged overall function. Notably, different signaling pathways were affected in the heart, including reduced expression of mitochondrial proteins, and elevated AKT, GSK3β, mTOR, MEK1/2 and ERK1/2 phosphorylation. Combined, our data demonstrate detrimental effects on skeletal and cardiac muscle in association with chronic administration of MKIs, although different mechanisms would seem to contribute to the cachectic phenotype in the two tissues.
Collapse
Affiliation(s)
- Joshua R Huot
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Alyson L Essex
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Maya Gutierrez
- Greenfield Central High School, Greenfield, IN 46140, USA.
| | - Rafael Barreto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Meijing Wang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - David L Waning
- Department of Cellular and Molecular Physiology, Penn State University, Hershey, PA 17033, USA.
| | - Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Otolaryngology-Head & Neck Surgery, Indiana Center for Musculoskeletal Health, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
8
|
Malentacchi F, Turrini I, Sorbi F, Projetto E, Castiglione F, Fambrini M, Petraglia F, Pillozzi S, Noci I. Pilot investigation of the mutation profile of PIK3CA/PTEN genes (PI3K pathway) in grade 3 endometrial cancer. Oncol Rep 2018; 41:1560-1574. [PMID: 30569174 PMCID: PMC6365709 DOI: 10.3892/or.2018.6939] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/09/2018] [Indexed: 12/31/2022] Open
Abstract
Endometrial cancer (EC) comprises a biological and clinical heterogeneous group of tumors. Several genetic alterations are involved in the development and progression of EC, and may be used for targeted therapy, particularly in patients with advanced-stage EC. In the present study, a combined procedure was developed based on polymerase chain reaction (PCR)-high resolution melting analysis (HRMA) and Sanger sequencing for the evaluation of somatic mutations in selected phosphoinositide 3-kinase (PI3K) catalytic subunit α (PIK3CA; exons 1, 9 and 21) and phosphatase and tensin homolog (PTEN; exons 5, 6, 7 and 8) exons. This combined procedure has the specificity and sensitivity of the two techniques, and overcomes their limitations. A pilot study was performed on 18 selected homogenous EC samples, of grade 3 endometrioid subtype (G3 EEC). First, the feasibility of the combined procedure was investigated to properly identify the presence of somatic mutations on PIK3CA and PTEN, the variations identified were analyzed using Catalogue of Somatic Mutations in Cancer, PolyPhen-2 and Mutation Taster software, and the frequency of mutations/variations was determined in the selected samples. The evaluation of mutational load revealed that the majority of the G3 EEC samples exhibited PIK3CA mutations (39%) and PTEN mutations (67%), and the majority of the samples (83%) had mutations in at least one of the two genes, and 33% had mutations in the two genes. The results of the present pilot study suggested that the cost-effective combined PCR-HRMA and Sanger sequencing procedure may be suitable for identification of PTEN and PIK3CA mutations in G3 EEC and that their frequency was consistent in G3 EEC, indicating that the PI3K pathway serves a pivotal function that may have potential for defining targeted therapy for the treatment of G3 EEC.
Collapse
Affiliation(s)
- Francesca Malentacchi
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, I‑50134 Florence, Italy
| | - Irene Turrini
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, I‑50134 Florence, Italy
| | - Flavia Sorbi
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, I‑50134 Florence, Italy
| | - Elisabetta Projetto
- Department of Surgery and Translational Medicine, Division of Pathological Anatomy, University of Florence, I‑50134 Florence, Italy
| | - Francesca Castiglione
- Department of Surgery and Translational Medicine, Division of Pathological Anatomy, University of Florence, I‑50134 Florence, Italy
| | - Massimiliano Fambrini
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, I‑50134 Florence, Italy
| | - Felice Petraglia
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, I‑50134 Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, I‑50134 Florence, Italy
| | - Ivo Noci
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, I‑50134 Florence, Italy
| |
Collapse
|
9
|
Dosil MA, Navaridas R, Mirantes C, Tarragona J, Eritja N, Felip I, Urdanibia I, Megino C, Domingo M, Santacana M, Gatius S, Piñol C, Barceló C, Maiques O, Macià A, Velasco A, Vaquero M, Matias-Guiu X, Dolcet X. Tumor suppressive function of E2F-1 on PTEN-induced serrated colorectal carcinogenesis. J Pathol 2018; 247:72-85. [DOI: 10.1002/path.5168] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 08/22/2018] [Accepted: 09/04/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Maria A Dosil
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Raúl Navaridas
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Cristina Mirantes
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Jordi Tarragona
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Núria Eritja
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Isidre Felip
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Izaskun Urdanibia
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Cristina Megino
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Mónica Domingo
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Maria Santacana
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Sònia Gatius
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Carme Piñol
- Department de Medicina; Universitat de Lleida-Institut de Recerca Biomèdica de Lleida (IRBLleida); Lleida Spain
| | - Carla Barceló
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Oscar Maiques
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Anna Macià
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Ana Velasco
- Department of Pathology and Molecular Genetics; Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLleida; Lleida Spain
| | - Marta Vaquero
- Department of Pathology and Molecular Genetics; Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLleida; Lleida Spain
| | - Xavier Matias-Guiu
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Xavier Dolcet
- Oncologic Pathology Group, Department de Ciències Mèdiques Bàsiques, Universitat de Lleida, Hospital Universitari Arnau de Vilanova; Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| |
Collapse
|
10
|
Ullman D, Dorn D, Rais-Bahrami S, Gordetsky J. Clinical Utility and Biologic Implications of Phosphatase and Tensin Homolog (PTEN) and ETS-related Gene (ERG) in Prostate Cancer. Urology 2017; 113:59-70. [PMID: 29225123 DOI: 10.1016/j.urology.2017.11.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 12/13/2022]
Abstract
Phosphatase and tensin homolog (PTEN) and ETS-related gene (ERG) mutations are commonly found in prostate cancer. Although mouse studies have demonstrated that PTEN and ERG cooperatively interact during tumorigenesis, human studies examining these genes have been inconclusive. A systematic PubMed search including original articles assessing the pathogenesis of PTEN and ERG in prostate cancer was performed. Studies examining ERG's prognostic significance have conflicting results. Studies examining PTEN and ERG simultaneously found these genes are likely to occur together, but cooperative tumorigenesis functions have not been conclusively established. PTEN mutations are associated with a range of prognostic features. However, the practical clinical utility of this information remains to be determined.
Collapse
Affiliation(s)
- David Ullman
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - David Dorn
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Soroush Rais-Bahrami
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL; Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Jennifer Gordetsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL; Department of Urology, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
11
|
Dosil MA, Mirantes C, Eritja N, Felip I, Navaridas R, Gatius S, Santacana M, Colàs E, Moiola C, Schoenenberger JA, Encinas M, Garí E, Matias-Guiu X, Dolcet X. Palbociclib has antitumour effects on Pten-
deficient endometrial neoplasias. J Pathol 2017; 242:152-164. [DOI: 10.1002/path.4896] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 02/08/2017] [Accepted: 03/09/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Maria Alba Dosil
- Oncologic Pathology Group, Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida; Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Cristina Mirantes
- Oncologic Pathology Group, Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida; Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Núria Eritja
- Oncologic Pathology Group, Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida; Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Isidre Felip
- Oncologic Pathology Group, Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida; Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Raúl Navaridas
- Oncologic Pathology Group, Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida; Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Sònia Gatius
- Oncologic Pathology Group, Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida; Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Maria Santacana
- Oncologic Pathology Group, Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida; Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Eva Colàs
- Biomedical Research Group in Gynaecology, Vall Hebron Research Institute (VHIR); Universitat Autònoma de Barcelona; Barcelona Spain
| | - Cristian Moiola
- Biomedical Research Group in Gynaecology, Vall Hebron Research Institute (VHIR); Universitat Autònoma de Barcelona; Barcelona Spain
| | - Joan Antoni Schoenenberger
- Department of Pharmacology, Hospital Universitari Arnau de Vilanova. Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Mario Encinas
- Departament de Medicina Experimental, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Eloi Garí
- Cell Cycle Group, Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
| | - Xavier Matias-Guiu
- Oncologic Pathology Group, Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida; Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| | - Xavier Dolcet
- Oncologic Pathology Group, Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida; Hospital Universitari Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, IRBLleida; Lleida Spain
- Centro de Investigación Biomédica en Red de Oncología (CIBERONC); Madrid Spain
| |
Collapse
|
12
|
Boswell BA, Korol A, West-Mays JA, Musil LS. Dual function of TGFβ in lens epithelial cell fate: implications for secondary cataract. Mol Biol Cell 2017; 28:907-921. [PMID: 28209733 PMCID: PMC5385940 DOI: 10.1091/mbc.e16-12-0865] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/06/2017] [Accepted: 02/07/2017] [Indexed: 12/29/2022] Open
Abstract
The most common vision-disrupting complication of cataract surgery is posterior capsule opacification (PCO; secondary cataract). PCO is caused by residual lens cells undergoing one of two very different cell fates: either transdifferentiating into myofibroblasts or maturing into lens fiber cells. Although TGFβ has been strongly implicated in lens cell fibrosis, the factors responsible for the latter process have not been identified. We show here for the first time that TGFβ can induce purified primary lens epithelial cells within the same culture to undergo differentiation into either lens fiber cells or myofibroblasts. Marker analysis confirmed that the two cell phenotypes were mutually exclusive. Blocking the p38 kinase pathway, either with direct inhibitors of the p38 MAP kinase or a small-molecule therapeutic that also inhibits the activation of p38, prevented TGFβ from inducing epithelial-myofibroblast transition and cell migration but did not prevent fiber cell differentiation. Rapamycin had the converse effect, linking MTOR signaling to induction of fiber cell differentiation by TGFβ. In addition to providing novel potential therapeutic strategies for PCO, our findings extend the so-called TGFβ paradox, in which TGFβ can induce two disparate cell fates, to a new epithelial disease state.
Collapse
Affiliation(s)
- Bruce A Boswell
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239
| | - Anna Korol
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Hamilton, ON L8N 3Z5, Canada
| | - Judith A West-Mays
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Hamilton, ON L8N 3Z5, Canada
| | - Linda S Musil
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239
| |
Collapse
|
13
|
Tuchen M, Wilisch-Neumann A, Daniel EA, Baldauf L, Pachow D, Scholz J, Angenstein F, Stork O, Kirches E, Mawrin C. Receptor tyrosine kinase inhibition by regorafenib/sorafenib inhibits growth and invasion of meningioma cells. Eur J Cancer 2017; 73:9-21. [PMID: 28082204 DOI: 10.1016/j.ejca.2016.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 09/28/2016] [Accepted: 12/06/2016] [Indexed: 12/19/2022]
Abstract
Systemic chemotherapeutic treatment for unresectable and/or aggressive meningiomas is still unsatisfying. PDGF receptor (PDGFR)-mediated activation of mitogenic signalling has been shown to be active in meningiomas. Therefore, we evaluate in vitro and in vivo the effects of inhibiting PDGFR using the clinically well-characterised tyrosine kinase inhibitors sorafenib or regorafenib in meningioma models. IOMM-Lee meningioma cells were used to assess cytotoxic effects, inhibition of proliferation, induction of apoptosis, as well as inhibition of migration and motility by sorafenib and regorafenib. Using an orthotopic mouse xenograft model, growth inhibition as monitored by magnetic resonance imaging, and overall survival of sorafenib- or regorafenib-treated mice compared with control animals was determined. Treatment of malignant IOMM-Lee cells resulted in significantly reduced cell survival and induction of apoptosis following regorafenib and sorafenib treatment. Western blots showed that both drugs target phosphorylation of p44/42 ERK via downregulation of the PDGFR. Both drugs additionally showed significant inhibition of cell motility and invasion. In vivo, mice with orthotopic meningioma xenografts showed a reduced volume (n.s.) of signal enhancement in MRI (mainly tumour) following sorafenib and regorafenib treatment. This was translated in a significantly increased overall survival time (p ≤ 0.05) for regorafenib-treated mice. Analyses of in vivo-grown tumours demonstrated again reduced PDGFR expression and expression/phosphorylation of p44/42. Sorafenib and regorafenib show antitumour activity in vitro and in vivo by targeting PDGFR and p44/42 ERK signalling.
Collapse
Affiliation(s)
- Marcus Tuchen
- Department of Neuropathology & Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke-University Magdeburg, and Center of Behavioral Brain Science, Magdeburg, Germany
| | - Annette Wilisch-Neumann
- Department of Neuropathology & Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke-University Magdeburg, and Center of Behavioral Brain Science, Magdeburg, Germany
| | - Evelyn A Daniel
- Department of Neuropathology & Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke-University Magdeburg, and Center of Behavioral Brain Science, Magdeburg, Germany
| | - Lisa Baldauf
- Department of Neuropathology & Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke-University Magdeburg, and Center of Behavioral Brain Science, Magdeburg, Germany
| | - Doreen Pachow
- Department of Neuropathology & Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke-University Magdeburg, and Center of Behavioral Brain Science, Magdeburg, Germany
| | - Johannes Scholz
- Department of Neuropathology & Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke-University Magdeburg, and Center of Behavioral Brain Science, Magdeburg, Germany
| | - Frank Angenstein
- DZNE, Department for Genetics & Molecular Neurobiology, Otto-von-Guericke-University Magdeburg, and Center of Behavioral Brain Science, Magdeburg, Germany
| | - Oliver Stork
- Institute of Biology, Department for Genetics & Molecular Neurobiology, Otto-von-Guericke-University Magdeburg, and Center of Behavioral Brain Science, Magdeburg, Germany
| | - Elmar Kirches
- Department of Neuropathology & Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke-University Magdeburg, and Center of Behavioral Brain Science, Magdeburg, Germany
| | - Christian Mawrin
- Department of Neuropathology & Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke-University Magdeburg, and Center of Behavioral Brain Science, Magdeburg, Germany.
| |
Collapse
|