1
|
Mehta D, Rajput K, Jain D, Bajaj A, Dasgupta U. Unveiling the Role of Mechanistic Target of Rapamycin Kinase (MTOR) Signaling in Cancer Progression and the Emergence of MTOR Inhibitors as Therapeutic Strategies. ACS Pharmacol Transl Sci 2024; 7:3758-3779. [PMID: 39698262 PMCID: PMC11650738 DOI: 10.1021/acsptsci.4c00530] [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: 09/02/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024]
Abstract
The mechanistic target of rapamycin kinase (MTOR) is pivotal for cell growth, metabolism, and survival. It functions through two distinct complexes, mechanistic TORC1 and mechanistic TORC2 (mTORC1 and mTORC2). These complexes function in the development and progression of cancer by regulating different cellular processes, such as protein synthesis, lipid metabolism, and glucose homeostasis. The mTORC1 complex senses nutrients and initiates proliferative signals, and mTORC2 is crucial for cell survival and cytoskeletal rearrangements. mTORC1 and mTORC2 have therefore emerged as potential targets for cancer treatment. Several mTOR inhibitors, including rapamycin and its analogs (rapalogs), primarily target mTORC1 and are effective for specific cancer types. However, these inhibitors often lead to resistance and limited long-term advantages due to the activation of survival pathways through feedback mechanisms. Researchers have created next-generation inhibitors targeting mTORC1 and mTORC2 and dual PI3K/mTOR inhibitors to address these difficulties. These inhibitors demonstrate enhanced anti-tumor effects by simultaneously disrupting multiple signaling pathways and show promise for improved and long-lasting therapies. However, development of resistance and adverse side effects remain a significant obstacle. Recent additions known as RapaLinks have emerged as a boon to counter drug-resistant cancer cells, as they are more potent and provide a more comprehensive blockade of mTOR signaling pathways. This Review combines current research findings and clinical insights to enhance our understanding of the crucial role of mTOR signaling in cancer biology and highlights the evolution of mTOR inhibitors as promising therapeutic approaches.
Collapse
Affiliation(s)
- Devashish Mehta
- Amity
Institute of Integrative Sciences and Health, Amity University Haryana, Panchgaon, Manesar, Gurgaon-122413, Haryana, India
| | - Kajal Rajput
- Amity
Institute of Integrative Sciences and Health, Amity University Haryana, Panchgaon, Manesar, Gurgaon-122413, Haryana, India
| | - Dolly Jain
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon
Expressway, Faridabad-121001, Haryana, India
| | - Avinash Bajaj
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon
Expressway, Faridabad-121001, Haryana, India
| | - Ujjaini Dasgupta
- Amity
Institute of Integrative Sciences and Health, Amity University Haryana, Panchgaon, Manesar, Gurgaon-122413, Haryana, India
| |
Collapse
|
2
|
Onaka GM, de Carvalho MR, Onaka PK, Barbosa CM, Martinez PF, de Oliveira-Junior SA. Exercise, mTOR Activation, and Potential Impacts on the Liver in Rodents. BIOLOGY 2024; 13:362. [PMID: 38927242 PMCID: PMC11201249 DOI: 10.3390/biology13060362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 06/28/2024]
Abstract
The literature offers a consensus on the association between exercise training (ET) protocols based on the adequate parameters of intensity and frequency, and several adaptive alterations in the liver. Indeed, regular ET can reverse glucose and lipid metabolism disorders, especially from aerobic modalities, which can decrease intrahepatic fat formation. In terms of molecular mechanisms, the regulation of hepatic fat formation would be directly related to the modulation of the mechanistic target of rapamycin (mTOR), which would be stimulated by insulin signaling and Akt activation, from the following three different primary signaling pathways: (I) growth factor, (II) energy/ATP-sensitive, and (III) amino acid-sensitive signaling pathways, respectively. Hyperactivation of the Akt/mTORC1 pathway induces lipogenesis by regulating the action of sterol regulatory element binding protein-1 (SREBP-1). Exercise training interventions have been associated with multiple metabolic and tissue benefits. However, it is worth highlighting that the mTOR signaling in the liver in response to exercise interventions remains unclear. Hepatic adaptive alterations seem to be most outstanding when sustained by chronic interventions or high-intensity exercise protocols.
Collapse
Affiliation(s)
- Giuliano Moreto Onaka
- Graduate Program in Health and Development in the Midwest Region, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil; (G.M.O.); (P.F.M.)
| | - Marianna Rabelo de Carvalho
- Graduate Program in Health and Development in the Midwest Region, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil; (G.M.O.); (P.F.M.)
| | - Patricia Kubalaki Onaka
- Graduate Program in Education and Health, State University of Mato Grosso do Sul, Dourados 79804-970, MS, Brazil
| | - Claudiane Maria Barbosa
- Graduate Program in Movement Sciences, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil;
| | - Paula Felippe Martinez
- Graduate Program in Health and Development in the Midwest Region, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil; (G.M.O.); (P.F.M.)
- Graduate Program in Movement Sciences, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil;
| | - Silvio Assis de Oliveira-Junior
- Graduate Program in Health and Development in the Midwest Region, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil; (G.M.O.); (P.F.M.)
- Graduate Program in Movement Sciences, Federal University of Mato Grosso do Sul—UFMS, Campo Grande 79070-900, MS, Brazil;
| |
Collapse
|
3
|
Ponzone L, Audrito V, Landi C, Moiso E, Levra Levron C, Ferrua S, Savino A, Vitale N, Gasparrini M, Avalle L, Vantaggiato L, Shaba E, Tassone B, Saoncella S, Orso F, Viavattene D, Marina E, Fiorilla I, Burrone G, Abili Y, Altruda F, Bini L, Deaglio S, Defilippi P, Menga A, Poli V, Porporato PE, Provero P, Raffaelli N, Riganti C, Taverna D, Cavallo F, Calautti E. RICTOR/mTORC2 downregulation in BRAF V600E melanoma cells promotes resistance to BRAF/MEK inhibition. Mol Cancer 2024; 23:105. [PMID: 38755661 PMCID: PMC11097536 DOI: 10.1186/s12943-024-02010-1] [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: 09/30/2023] [Accepted: 04/26/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND The main drawback of BRAF/MEK inhibitors (BRAF/MEKi)-based targeted therapy in the management of BRAF-mutated cutaneous metastatic melanoma (MM) is the development of therapeutic resistance. We aimed to assess in this context the role of mTORC2, a signaling complex defined by the presence of the essential RICTOR subunit, regarded as an oncogenic driver in several tumor types, including MM. METHODS After analyzing The Cancer Genome Atlas MM patients' database to explore both overall survival and molecular signatures as a function of intra-tumor RICTOR levels, we investigated the effects of RICTOR downregulation in BRAFV600E MM cell lines on their response to BRAF/MEKi. We performed proteomic screening to identify proteins modulated by changes in RICTOR expression, and Seahorse analysis to evaluate the effects of RICTOR depletion on mitochondrial respiration. The combination of BRAFi with drugs targeting proteins and processes emerged in the proteomic screening was carried out on RICTOR-deficient cells in vitro and in a xenograft setting in vivo. RESULTS Low RICTOR levels in BRAF-mutated MM correlate with a worse clinical outcome. Gene Set Enrichment Analysis of low-RICTOR tumors display gene signatures suggestive of activation of the mitochondrial Electron Transport Chain (ETC) energy production. RICTOR-deficient BRAFV600E cells are intrinsically tolerant to BRAF/MEKi and anticipate the onset of resistance to BRAFi upon prolonged drug exposure. Moreover, in drug-naïve cells we observed a decline in RICTOR expression shortly after BRAFi exposure. In RICTOR-depleted cells, both mitochondrial respiration and expression of nicotinamide phosphoribosyltransferase (NAMPT) are enhanced, and their pharmacological inhibition restores sensitivity to BRAFi. CONCLUSIONS Our work unveils an unforeseen tumor-suppressing role for mTORC2 in the early adaptation phase of BRAFV600E melanoma cells to targeted therapy and identifies the NAMPT-ETC axis as a potential therapeutic vulnerability of low RICTOR tumors. Importantly, our findings indicate that the evaluation of intra-tumor RICTOR levels has a prognostic value in metastatic melanoma and may help to guide therapeutic strategies in a personalized manner.
Collapse
Affiliation(s)
- Luca Ponzone
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Valentina Audrito
- Department of Science and Technological Innovation, University of Piemonte Orientale, Alessandria, 15121, Italy
| | - Claudia Landi
- Functional Proteomic Section, Department of Life Sciences, University of Siena, Siena, 53100, Italy
| | - Enrico Moiso
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Chiara Levra Levron
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Life Sciences and Systems Biology, University of Turin, Turin, 10126, Italy
| | - Sara Ferrua
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Aurora Savino
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Nicoletta Vitale
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Massimiliano Gasparrini
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, 60131, Italy
| | - Lidia Avalle
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Department of Science and Technological Innovation, University of Piemonte Orientale, Alessandria, 15121, Italy
| | - Lorenza Vantaggiato
- Functional Proteomic Section, Department of Life Sciences, University of Siena, Siena, 53100, Italy
| | - Enxhi Shaba
- Functional Proteomic Section, Department of Life Sciences, University of Siena, Siena, 53100, Italy
| | - Beatrice Tassone
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Department of Personal Care, dsm-firmenich, Kaiseraugst, 4303, Switzerland
| | - Stefania Saoncella
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Francesca Orso
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Daniele Viavattene
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Eleonora Marina
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Irene Fiorilla
- Department of Science and Technological Innovation, University of Piemonte Orientale, Alessandria, 15121, Italy
| | - Giulia Burrone
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- Department of Clinical and Biological Sciences, University of Turin, Turin, 10124, Italy
| | - Youssef Abili
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
- GenomeUp, Rome, 00144, Italy
| | - Fiorella Altruda
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Luca Bini
- Functional Proteomic Section, Department of Life Sciences, University of Siena, Siena, 53100, Italy
| | - Silvia Deaglio
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Medical Sciences, University of Turin, Turin, 10124, Italy
| | - Paola Defilippi
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Alessio Menga
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Valeria Poli
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Paolo Ettore Porporato
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Paolo Provero
- Neuroscience Department "Rita Levi Montalcini", University of Turin, Turin, 10126, Italy
| | - Nadia Raffaelli
- Department of Agriculture, Food and Environmental Sciences, Polytechnic University of Marche, Ancona, 60131, Italy
| | - Chiara Riganti
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Oncology, University of Turin, Turin, 10124, Italy
| | - Daniela Taverna
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Federica Cavallo
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy
| | - Enzo Calautti
- Molecular Biotechnology Center "Guido Tarone", University of Turin, Turin, 10126, Italy.
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, 10126, Italy.
| |
Collapse
|
4
|
Krencz I, Sztankovics D, Sebestyén A, Pápay J, Dankó T, Moldvai D, Lutz E, Khoor A. RICTOR amplification is associated with Rictor membrane staining and does not correlate with PD-L1 expression in lung squamous cell carcinoma. Pathol Oncol Res 2024; 30:1611593. [PMID: 38706776 PMCID: PMC11066283 DOI: 10.3389/pore.2024.1611593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/04/2024] [Indexed: 05/07/2024]
Abstract
RICTOR gene, which encodes the scaffold protein of mTORC2, can be amplified in various tumor types, including squamous cell carcinoma (SCC) of the lung. RICTOR amplification can lead to hyperactivation of mTORC2 and may serve as a targetable genetic alteration, including in lung SCC patients with no PD-L1 expression who are not expected to benefit from immune checkpoint inhibitor therapy. This study aimed to compare RICTOR amplification detected by fluorescence in situ hybridization (FISH) with Rictor and PD-L1 protein expression detected by immunohistochemistry (IHC) in SCC of the lung. The study was complemented by analysis of the publicly available Lung Squamous Cell Carcinoma (TCGA, Firehose legacy) dataset. RICTOR amplification was observed in 20% of our cases and 16% of the lung SCC cases of the TCGA dataset. Rictor and PD-L1 expression was seen in 74% and 44% of the cases, respectively. Rictor IHC showed two staining patterns: membrane staining (16% of the cases) and cytoplasmic staining (58% of the cases). Rictor membrane staining predicted RICTOR amplification as detected by FISH with high specificity (95%) and sensitivity (70%). We did not find any correlation between RICTOR amplification and PD-L1 expression; RICTOR amplification was detected in 18% and 26% of PD-L1 positive and negative cases, respectively. The TCGA dataset analysis showed similar results; RICTOR copy number correlated with Rictor mRNA and protein expression but showed no association with PD-L1 mRNA and protein expression. In conclusion, the correlation between RICTOR amplification and Rictor membrane staining suggests that the latter can potentially be used as a surrogate marker to identify lung SCC cases with RICTOR amplification. Since a significant proportion of PD-L1 negative SCC cases harbor RICTOR amplification, analyzing PD-L1 negative tumors by RICTOR FISH or Rictor IHC can help select patients who may benefit from mTORC2 inhibitor therapy.
Collapse
Affiliation(s)
- Ildikó Krencz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Dániel Sztankovics
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Judit Pápay
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Titanilla Dankó
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Dorottya Moldvai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Elmar Lutz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, United States
| | - Andras Khoor
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
5
|
Sztankovics D, Moldvai D, Petővári G, Dankó T, Szalai F, Miyaura R, Varga V, Nagy N, Papp G, Pápay J, Krencz I, Sebestyén A. mTOR hyperactivity and RICTOR amplification as targets for personalized treatments in malignancies. Pathol Oncol Res 2024; 30:1611643. [PMID: 38515456 PMCID: PMC10954904 DOI: 10.3389/pore.2024.1611643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
The increasing knowledge of molecular alterations in malignancies, including mutations and regulatory failures in the mTOR (mechanistic target of rapamycin) signaling pathway, highlights the importance of mTOR hyperactivity as a validated target in common and rare malignancies. This review summarises recent findings on the characterization and prognostic role of mTOR kinase complexes (mTORC1 and mTORC2) activity regarding differences in their function, structure, regulatory mechanisms, and inhibitor sensitivity. We have recently identified new tumor types with RICTOR (rapamycin-insensitive companion of mTOR) amplification and associated mTORC2 hyperactivity as useful potential targets for developing targeted therapies in lung cancer and other newly described malignancies. The activity of mTOR complexes is recommended to be assessed and considered in cancers before mTOR inhibitor therapy, as current first-generation mTOR inhibitors (rapamycin and analogs) can be ineffective in the presence of mTORC2 hyperactivity. We have introduced and proposed a marker panel to determine tissue characteristics of mTOR activity in biopsy specimens, patient materials, and cell lines. Ongoing phase trials of new inhibitors and combination therapies are promising in advanced-stage patients selected by genetic alterations, molecular markers, and/or protein expression changes in the mTOR signaling pathway. Hopefully, the summarized results, our findings, and the suggested characterization of mTOR activity will support therapeutic decisions.
Collapse
|
6
|
Szalai F, Sztankovics D, Krencz I, Moldvai D, Pápay J, Sebestyén A, Khoor A. Rictor-A Mediator of Progression and Metastasis in Lung Cancer. Cancers (Basel) 2024; 16:543. [PMID: 38339294 PMCID: PMC10854599 DOI: 10.3390/cancers16030543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Lung carcinoma is one of the most common cancer types for both men and women. Despite recent breakthroughs in targeted therapy and immunotherapy, it is characterized by a high metastatic rate, which can significantly affect quality of life and prognosis. Rictor (encoded by the RICTOR gene) is known as a scaffold protein for the multiprotein complex mTORC2. Among its diverse roles in regulating essential cellular functions, mTORC2 also facilitates epithelial-mesenchymal transition and metastasis formation. Amplification of the RICTOR gene and subsequent overexpression of the Rictor protein can result in the activation of mTORC2, which promotes cell survival and migration. Based on recent studies, RICTOR amplification or Rictor overexpression can serve as a marker for mTORC2 activation, which in turn provides a promising druggable target. Although selective inhibitors of Rictor and the Rictor-mTOR association are only in a preclinical phase, they seem to be potent novel approaches to reduce tumor cell migration and metastasis formation. Here, we summarize recent advances that support an important role for Rictor and mTORC2 as potential therapeutic targets in the treatment of lung cancer. This is a traditional (narrative) review based on Pubmed and Google Scholar searches for the following keywords: Rictor, RICTOR amplification, mTORC2, Rictor complexes, lung cancer, metastasis, progression, mTOR inhibitors.
Collapse
Affiliation(s)
- Fatime Szalai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Dániel Sztankovics
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Ildikó Krencz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Dorottya Moldvai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Judit Pápay
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Andras Khoor
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| |
Collapse
|
7
|
Cheng KP, Shen WX, Jiang YY, Chen Y, Chen YZ, Tan Y. Deep learning of 2D-Restructured gene expression representations for improved low-sample therapeutic response prediction. Comput Biol Med 2023; 164:107245. [PMID: 37480677 DOI: 10.1016/j.compbiomed.2023.107245] [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: 04/26/2023] [Revised: 06/27/2023] [Accepted: 07/07/2023] [Indexed: 07/24/2023]
Abstract
Clinical outcome prediction is important for stratified therapeutics. Machine learning (ML) and deep learning (DL) methods facilitate therapeutic response prediction from transcriptomic profiles of cells and clinical samples. Clinical transcriptomic DL is challenged by the low-sample sizes (34-286 subjects), high-dimensionality (up to 21,653 genes) and unordered nature of clinical transcriptomic data. The established methods rely on ML algorithms at accuracy levels of 0.6-0.8 AUC/ACC values. Low-sample DL algorithms are needed for enhanced prediction capability. Here, an unsupervised manifold-guided algorithm was employed for restructuring transcriptomic data into ordered image-like 2D-representations, followed by efficient DL of these 2D-representations with deep ConvNets. Our DL models significantly outperformed the state-of-the-art (SOTA) ML models on 82% of 17 low-sample benchmark datasets (53% with >0.05 AUC/ACC improvement). They are more robust than the SOTA models in cross-cohort prediction tasks, and in identifying robust biomarkers and response-dependent variational patterns consistent with experimental indications.
Collapse
Affiliation(s)
- Kai Ping Cheng
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, PR China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, PR China
| | - Wan Xiang Shen
- Bioinformatics and Drug Design Group, Department of Pharmacy, Center for Computational Science and Engineering, National University of Singapore, 117543, Singapore
| | - Yu Yang Jiang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, PR China
| | - Yan Chen
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, PR China
| | - Yu Zong Chen
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, PR China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, PR China.
| | - Ying Tan
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, PR China; The Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, PR China; Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen, 518110, PR China.
| |
Collapse
|
8
|
Githaka JM, Pirayeshfard L, Goping IS. Cancer invasion and metastasis: Insights from murine pubertal mammary gland morphogenesis. Biochim Biophys Acta Gen Subj 2023; 1867:130375. [PMID: 37150225 DOI: 10.1016/j.bbagen.2023.130375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/20/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Cancer invasion and metastasis accounts for the majority of cancer related mortality. A better understanding of the players that drive the aberrant invasion and migration of tumors cells will provide critical targets to inhibit metastasis. Postnatal pubertal mammary gland morphogenesis is characterized by highly proliferative, invasive, and migratory normal epithelial cells. Identifying the molecular regulators of pubertal gland development is a promising strategy since tumorigenesis and metastasis is postulated to be a consequence of aberrant reactivation of developmental stages. In this review, we summarize the pubertal morphogenesis regulators that are involved in cancer metastasis and revisit pubertal mammary gland transcriptome profiling to uncover both known and unknown metastasis genes. Our updated list of pubertal morphogenesis regulators shows that most are implicated in invasion and metastasis. This review highlights molecular linkages between development and metastasis and provides a guide for exploring novel metastatic drivers.
Collapse
Affiliation(s)
- John Maringa Githaka
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Leila Pirayeshfard
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ing Swie Goping
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Department of Oncology, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
9
|
Zhang X, Evans TD, Chen S, Sergin I, Stitham J, Jeong SJ, Rodriguez-Velez A, Yeh YS, Park A, Jung IH, Diwan A, Schilling JD, Rom O, Yurdagul A, Epelman S, Cho J, Lodhi IJ, Mittendorfer B, Razani B. Loss of Macrophage mTORC2 Drives Atherosclerosis via FoxO1 and IL-1β Signaling. Circ Res 2023; 133:200-219. [PMID: 37350264 PMCID: PMC10527041 DOI: 10.1161/circresaha.122.321542] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 06/12/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND The mTOR (mechanistic target of rapamycin) pathway is a complex signaling cascade that regulates cellular growth, proliferation, metabolism, and survival. Although activation of mTOR signaling has been linked to atherosclerosis, its direct role in lesion progression and in plaque macrophages remains poorly understood. We previously demonstrated that mTORC1 (mTOR complex 1) activation promotes atherogenesis through inhibition of autophagy and increased apoptosis in macrophages. METHODS Using macrophage-specific Rictor- and mTOR-deficient mice, we now dissect the distinct functions of mTORC2 pathways in atherogenesis. RESULTS In contrast to the atheroprotective effect seen with blockade of macrophage mTORC1, macrophage-specific mTORC2-deficient mice exhibit an atherogenic phenotype, with larger, more complex lesions and increased cell death. In cultured macrophages, we show that mTORC2 signaling inhibits the FoxO1 (forkhead box protein O1) transcription factor, leading to suppression of proinflammatory pathways, especially the inflammasome/IL (interleukin)-1β response, a key mediator of vascular inflammation and atherosclerosis. In addition, administration of FoxO1 inhibitors efficiently rescued the proinflammatory response caused by mTORC2 deficiency both in vitro and in vivo. Interestingly, collective deletion of macrophage mTOR, which ablates mTORC1- and mTORC2-dependent pathways, leads to minimal change in plaque size or complexity, reflecting the balanced yet opposing roles of these signaling arms. CONCLUSIONS Our data provide the first mechanistic details of macrophage mTOR signaling in atherosclerosis and suggest that therapeutic measures aimed at modulating mTOR need to account for its dichotomous functions.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Trent D. Evans
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Sunny Chen
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Ismail Sergin
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Jeremiah Stitham
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | | | - Yu-Sheng Yeh
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Arick Park
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - In-Hyuk Jung
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Abhinav Diwan
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| | - Joel D. Schilling
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology and Department of Molecular and Cellular Physiology, Louisiana State University, Shreveport, LA
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology and Department of Molecular and Cellular Physiology, Louisiana State University, Shreveport, LA
| | - Slava Epelman
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac Center, Toronto General Hospital Research Institute, University Health Network and University of Toronto, Toronto, Canada
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Irfan J. Lodhi
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Bettina Mittendorfer
- Division of Geriatrics and Nutritional Science, and Washington University School of Medicine, St Louis, MO, USA
| | - Babak Razani
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Pittsburgh VA Medical Center, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| |
Collapse
|
10
|
Ezhilarasan D, Najimi M. Deciphering the possible reciprocal loop between hepatic stellate cells and cancer cells in the tumor microenvironment of the liver. Crit Rev Oncol Hematol 2023; 182:103902. [PMID: 36621514 DOI: 10.1016/j.critrevonc.2022.103902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/07/2023] Open
Abstract
Activated hepatic stellate cells (HSCs)/myofibroblasts are the important sources of cancer-associated fibroblasts in the liver tumor microenvironment (TME). The crosstalk between activated HSCs and tumor cells mediates HCC progression, metastasis, tumor cell survival, angiogenesis and chemoresistance. In TME, HCC cells secrete various soluble factors responsible for the phenotypic activation of quiescent HSCs. Tumor cells use activated HSC-derived extracellular matrix (ECM) for migration and invasion. Further, in liver TME, activated HSCs and sinusoidal endothelial cells engage in a crosstalk that causes the secretion of angiogenesis and metastasis-related growth factors and cytokines. Activated HSCs and immune cells crosstalk to decrease immune surveillance in the liver TME by increasing the population of T regulatory cells and M2 macrophages or myeloid-derived suppressor cells. Thus, HSCs play a vital role in liver TME cell interactions. Therefore, a deep understanding of HSCs activation and their crosstalk with cancer and immune cells in TME may lead to the development of novel therapeutic strategies to target HCC.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India.
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels 1200, Belgium
| |
Collapse
|
11
|
Gargalionis AN, Papavassiliou KA, Basdra EK, Papavassiliou AG. mTOR Signaling Components in Tumor Mechanobiology. Int J Mol Sci 2022; 23:1825. [PMID: 35163745 PMCID: PMC8837098 DOI: 10.3390/ijms23031825] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a central signaling hub that integrates networks of nutrient availability, cellular metabolism, and autophagy in eukaryotic cells. mTOR kinase, along with its upstream regulators and downstream substrates, is upregulated in most human malignancies. At the same time, mechanical forces from the tumor microenvironment and mechanotransduction promote cancer cells' proliferation, motility, and invasion. mTOR signaling pathway has been recently found on the crossroads of mechanoresponsive-induced signaling cascades to regulate cell growth, invasion, and metastasis in cancer cells. In this review, we examine the emerging association of mTOR signaling components with certain protein tools of tumor mechanobiology. Thereby, we highlight novel mechanisms of mechanotransduction, which regulate tumor progression and invasion, as well as mechanisms related to the therapeutic efficacy of antitumor drugs.
Collapse
Affiliation(s)
- Antonios N. Gargalionis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (E.K.B.)
- Department of Biopathology, Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Kostas A. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (E.K.B.)
| | - Efthimia K. Basdra
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (E.K.B.)
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (E.K.B.)
| |
Collapse
|
12
|
Joechle K, Jumaa H, Thriene K, Hellerbrand C, Kulemann B, Fichtner-Feigl S, Lang SA, Guenzle J. Dual Inhibition of mTORC1/2 Reduces Migration of Cholangiocarcinoma Cells by Regulation of Matrixmetalloproteinases. Front Cell Dev Biol 2022; 9:785979. [PMID: 35096817 PMCID: PMC8793831 DOI: 10.3389/fcell.2021.785979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/17/2021] [Indexed: 12/30/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a rare but highly aggressive tumor entity for which systemic therapies only showed limited efficacy so far. As OSI-027—a dual kinase inhibitor targeting both mTOR complexes, mTORC1 and mTORC2 - showed improved anti-cancer effects, we sought to evaluate its impact on the migratory and metastatic capacity of CCA cells in vitro. We found that treatment with OSI-027 leads to reduced cell mobility and migration as well as a reduced surviving fraction in colony-forming ability. While neither cell viability nor proliferation rate was affected, OSI-027 decreased the expression of MMP2 and MMP9. Moreover, survival as well as anti-apoptotic signaling was impaired upon the use of OSI-027 as determined by AKT and MAPK blotting. Dual targeting of mTORC1/2 might therefore be a viable option for anti-neoplastic therapy in CCA.
Collapse
Affiliation(s)
- Katharina Joechle
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany.,Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Huda Jumaa
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Kerstin Thriene
- Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Birte Kulemann
- Department of Surgery, University Medical Center Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany.,Comprehensive Cancer Center Freiburg-CCCF, Medical Center-University, Freiburg, Germany
| | - Sven A Lang
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany.,Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Jessica Guenzle
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
13
|
Karagianni F, Pavlidis A, Malakou LS, Piperi C, Papadavid E. Predominant Role of mTOR Signaling in Skin Diseases with Therapeutic Potential. Int J Mol Sci 2022; 23:ijms23031693. [PMID: 35163615 PMCID: PMC8835793 DOI: 10.3390/ijms23031693] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/30/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
The serine/threonine kinase mechanistic target of rapamycin (mTOR) plays a pivotal role in the regulation of cell proliferation, survival, and motility in response to availability of energy and nutrients as well as mitogens. The mTOR signaling axis regulates important biological processes, including cellular growth, metabolism, and survival in many tissues. In the skin, dysregulation of PI3K/AKT/mTOR pathway may lead to severe pathological conditions characterized by uncontrolled proliferation and inflammation, including skin hyperproliferative as well as malignant diseases. Herein, we provide an update on the current knowledge regarding the pathogenic implication of the mTOR pathway in skin diseases with inflammatory features (such as psoriasis, atopic dermatitis, pemphigus, and acne) and malignant characteristics (such as cutaneous T cell lymphoma and melanoma) while we critically discuss current and future perspectives for therapeutic targeting of mTOR axis in clinical practice.
Collapse
Affiliation(s)
- Fani Karagianni
- National Center of Rare Diseases—Cutaneous Lymphoma, Second Department of Dermatology and Venereal Diseases, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (F.K.); (A.P.); (E.P.)
| | - Antreas Pavlidis
- National Center of Rare Diseases—Cutaneous Lymphoma, Second Department of Dermatology and Venereal Diseases, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (F.K.); (A.P.); (E.P.)
| | - Lina S. Malakou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Correspondence:
| | - Evangelia Papadavid
- National Center of Rare Diseases—Cutaneous Lymphoma, Second Department of Dermatology and Venereal Diseases, Attikon University General Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece; (F.K.); (A.P.); (E.P.)
| |
Collapse
|
14
|
Joechle K, Guenzle J, Hellerbrand C, Strnad P, Cramer T, Neumann UP, Lang SA. Role of mammalian target of rapamycin complex 2 in primary and secondary liver cancer. World J Gastrointest Oncol 2021; 13:1632-1647. [PMID: 34853640 PMCID: PMC8603445 DOI: 10.4251/wjgo.v13.i11.1632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/30/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) acts in two structurally and functionally distinct protein complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Upon deregulation, activated mTOR signaling is associated with multiple processes involved in tumor growth and metastasis. Compared with mTORC1, much less is known about mTORC2 in cancer, mainly because of the unavailability of a selective inhibitor. However, existing data suggest that mTORC2 with its two distinct subunits Rictor and mSin1 might play a more important role than assumed so far. It is one of the key effectors of the PI3K/AKT/mTOR pathway and stimulates cell growth, cell survival, metabolism, and cytoskeletal organization. It is not only implicated in tumor progression, metastasis, and the tumor microenvironment but also in resistance to therapy. Rictor, the central subunit of mTORC2, was found to be upregulated in different kinds of cancers and is associated with advanced tumor stages and a bad prognosis. Moreover, AKT, the main downstream regulator of mTORC2/Rictor, is one of the most highly activated proteins in cancer. Primary and secondary liver cancer are major problems for current cancer therapy due to the lack of specific medical treatment, emphasizing the need for further therapeutic options. This review, therefore, summarizes the role of mTORC2/Rictor in cancer, with special focus on primary liver cancer but also on liver metastases.
Collapse
Affiliation(s)
- Katharina Joechle
- Department of General, Visceral and Transplantation Surgery, University Hospital Rheinisch-Westfälisch Technische Hochschule Aachen, Aachen 52074, Germany
| | - Jessica Guenzle
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Pavel Strnad
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälisch Technische Hochschule Aachen, Aachen 52074, Germany
| | - Thorsten Cramer
- Department of General, Visceral and Transplantation Surgery, University Hospital Rheinisch-Westfälisch Technische Hochschule Aachen, Aachen 52074, Germany
| | - Ulf Peter Neumann
- Department of General, Visceral and Transplantation Surgery, University Hospital Rheinisch-Westfälisch Technische Hochschule Aachen, Aachen 52074, Germany
| | - Sven Arke Lang
- Department of General, Visceral and Transplantation Surgery, University Hospital Rheinisch-Westfälisch Technische Hochschule Aachen, Aachen 52074, Germany
| |
Collapse
|
15
|
RICTOR Affects Melanoma Tumorigenesis and Its Resistance to Targeted Therapy. Biomedicines 2021; 9:biomedicines9101498. [PMID: 34680615 PMCID: PMC8533235 DOI: 10.3390/biomedicines9101498] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 12/22/2022] Open
Abstract
The network defined by phosphatidylinositol-3-kinase (PI3K), AKT, and mammalian target of rapamycin (mTOR) plays a major role in melanoma oncogenesis and has been implicated in BRAF inhibitor resistance. The central role of RICTOR (rapamycin-insensitive companion of mTOR) in this pathway has only recently begun to be unraveled. In the present study, we assessed the role of mTORC2/RICTOR in BRAF-mutated melanomas and their resistance to BRAF inhibition. We showed that RICTOR was significantly overexpressed in melanoma and associated with bad prognoses. RICTOR overexpression stimulated melanoma-initiating cells (MICs) with ‘stemness’ properties. We also showed that RICTOR contributed to melanoma resistance to BRAF inhibitors and rendered the cells very sensitive to mTORC2 inhibition. We highlighted a connection between mTORC2/RICTOR and STAT3 in resistant cells and revealed an interaction between RAS and RICTOR in resistant melanoma, which, when disrupted, impeded the proliferation of resistant cells. Therefore, as a key signaling node, RICTOR contributes to BRAF-dependent melanoma development and resistance to therapy and, as such, is a valuable therapeutic target in melanoma.
Collapse
|
16
|
Kumar S, Sánchez-Álvarez M, Lolo FN, Trionfetti F, Strippoli R, Cordani M. Autophagy and the Lysosomal System in Cancer. Cells 2021; 10:cells10102752. [PMID: 34685734 PMCID: PMC8534995 DOI: 10.3390/cells10102752] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 12/19/2022] Open
Abstract
Autophagy and the lysosomal system, together referred to as the autophagolysosomal system, is a cellular quality control network which maintains cellular health and homeostasis by removing cellular waste including protein aggregates, damaged organelles, and invading pathogens. As such, the autophagolysosomal system has roles in a variety of pathophysiological disorders, including cancer, neurological disorders, immune- and inflammation-related diseases, and metabolic alterations, among others. The autophagolysosomal system is controlled by TFEB, a master transcriptional regulator driving the expression of multiple genes, including autophagoly sosomal components. Importantly, Reactive Oxygen Species (ROS) production and control are key aspects of the physiopathological roles of the autophagolysosomal system, and may hold a key for synergistic therapeutic interventions. In this study, we reviewed our current knowledge on the biology and physiopathology of the autophagolysosomal system, and its potential for therapeutic intervention in cancer.
Collapse
Affiliation(s)
- Suresh Kumar
- Autophagy Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
- Correspondence: (S.K.); (R.S.)
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation & Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain; (M.S.-Á.); (F.-N.L.)
| | - Fidel-Nicolás Lolo
- Mechanoadaptation & Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain; (M.S.-Á.); (F.-N.L.)
| | - Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy;
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| | - Raffaele Strippoli
- Mechanoadaptation & Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain; (M.S.-Á.); (F.-N.L.)
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy;
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
- Correspondence: (S.K.); (R.S.)
| | | |
Collapse
|
17
|
Drexler K, Schmidt KM, Jordan K, Federlin M, Milenkovic VM, Liebisch G, Artati A, Schmidl C, Madej G, Tokarz J, Cecil A, Jagla W, Haerteis S, Aung T, Wagner C, Kolodziejczyk M, Heinke S, Stanton EH, Schwertner B, Riegel D, Wetzel CH, Buchalla W, Proescholdt M, Klein CA, Berneburg M, Schlitt HJ, Brabletz T, Ziegler C, Parkinson EK, Gaumann A, Geissler EK, Adamski J, Haferkamp S, Mycielska ME. Cancer-associated cells release citrate to support tumour metastatic progression. Life Sci Alliance 2021; 4:e202000903. [PMID: 33758075 PMCID: PMC7994318 DOI: 10.26508/lsa.202000903] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
Citrate is important for lipid synthesis and epigenetic regulation in addition to ATP production. We have previously reported that cancer cells import extracellular citrate via the pmCiC transporter to support their metabolism. Here, we show for the first time that citrate is supplied to cancer by cancer-associated stroma (CAS) and also that citrate synthesis and release is one of the latter's major metabolic tasks. Citrate release from CAS is controlled by cancer cells through cross-cellular communication. The availability of citrate from CAS regulated the cytokine profile, metabolism and features of cellular invasion. Moreover, citrate released by CAS is involved in inducing cancer progression especially enhancing invasiveness and organ colonisation. In line with the in vitro observations, we show that depriving cancer cells of citrate using gluconate, a specific inhibitor of pmCiC, significantly reduced the growth and metastatic spread of human pancreatic cancer cells in vivo and muted stromal activation and angiogenesis. We conclude that citrate is supplied to tumour cells by CAS and citrate uptake plays a significant role in cancer metastatic progression.
Collapse
Affiliation(s)
- Konstantin Drexler
- Department of Dermatology, University Medical Centre, Regensburg, Germany
| | | | - Katrin Jordan
- Department of Surgery, University Medical Center, Regensburg, Germany
| | - Marianne Federlin
- Department of Conservative Dentistry and Periodontology, University Medical Center, Regensburg, Germany
| | - Vladimir M Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, Regensburg, Germany
| | - Anna Artati
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Christian Schmidl
- Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Gregor Madej
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Regensburg, Germany
| | - Janina Tokarz
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Alexander Cecil
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
| | - Wolfgang Jagla
- Institute of Pathology, Kaufbeuren-Ravensburg, Kaufbeuren, Germany
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | - Thiha Aung
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
- Center of Plastic, Aesthetic, Hand and Reconstructive Surgery, University of Regensburg, Regensburg, Germany
| | - Christine Wagner
- Department of Surgery, University Medical Center, Regensburg, Germany
| | | | - Stefanie Heinke
- Department of Surgery, University Medical Center, Regensburg, Germany
| | - Evan H Stanton
- Department of Surgery, University Medical Center, Regensburg, Germany
| | - Barbara Schwertner
- Department of Dermatology, University Medical Centre, Regensburg, Germany
| | - Dania Riegel
- Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Christian H Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Wolfgang Buchalla
- Department of Conservative Dentistry and Periodontology, University Medical Center, Regensburg, Germany
| | - Martin Proescholdt
- Department of Neurosurgery, University Hospital Regensburg, Regensburg, Germany
| | - Christoph A Klein
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Medical Centre, Regensburg, Germany
| | - Hans J Schlitt
- Department of Surgery, University Medical Center, Regensburg, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Friedrich-Alexander-University Erlangen, Erlangen, Germany
| | - Christine Ziegler
- Department of Structural Biology, Institute of Biophysics and Physical Biochemistry, University of Regensburg, Regensburg, Germany
| | - Eric K Parkinson
- Centre for Immunobiology and Regenerative Medicine, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, UK
| | - Andreas Gaumann
- Institute of Pathology, Kaufbeuren-Ravensburg, Kaufbeuren, Germany
| | - Edward K Geissler
- Department of Surgery, University Medical Center, Regensburg, Germany
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Munich, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Maria E Mycielska
- Department of Surgery, University Medical Center, Regensburg, Germany
| |
Collapse
|
18
|
Guenzle J, Akasaka H, Joechle K, Reichardt W, Venkatasamy A, Hoeppner J, Hellerbrand C, Fichtner-Feigl S, Lang SA. Pharmacological Inhibition of mTORC2 Reduces Migration and Metastasis in Melanoma. Int J Mol Sci 2020; 22:ijms22010030. [PMID: 33375117 PMCID: PMC7792954 DOI: 10.3390/ijms22010030] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Despite recent advances in therapy, liver metastasis from melanoma is still associated with poor prognosis. Although targeting the mTOR signaling pathway exerts potent anti-tumor activity, little is known about specific mTORC2 inhibition regarding liver metastasis. Using the novel mTORC2 specific inhibitor JR-AB2-011, we show significantly reduced migration and invasion capacity by impaired activation of MMP2 in melanoma cells. In addition, blockade of mTORC2 induces cell death by non-apoptotic pathways and reduces tumor cell proliferation rate dose-dependently. Furthermore, a significant reduction of liver metastasis was detected in a syngeneic murine metastasis model upon therapy with JR-AB2-011 as determined by in vivo imaging and necropsy. Hence, our study for the first time highlights the impact of the pharmacological blockade of mTORC2 as a potent novel anti-cancer approach for liver metastasis from melanoma.
Collapse
Affiliation(s)
- Jessica Guenzle
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.G.); (H.A.); (K.J.); (J.H.); (S.F.-F.)
| | - Harue Akasaka
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.G.); (H.A.); (K.J.); (J.H.); (S.F.-F.)
| | - Katharina Joechle
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.G.); (H.A.); (K.J.); (J.H.); (S.F.-F.)
| | - Wilfried Reichardt
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany;
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Radiology Medical Physics, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Killianstrasse 5a, 79106 Freiburg, Germany;
| | - Aina Venkatasamy
- Department of Radiology Medical Physics, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Killianstrasse 5a, 79106 Freiburg, Germany;
- Service de Radiologie 1, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, 1 Avenue Molière, 67098 Strasbourg, France
- Laboratory Stress Response and Innovative Therapies “Streinth”, Inserm IRFAC UMR_S1113, Université de Strasbourg, 67098 Strasbourg, France
| | - Jens Hoeppner
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.G.); (H.A.); (K.J.); (J.H.); (S.F.-F.)
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich–Alexander University Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany;
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.G.); (H.A.); (K.J.); (J.H.); (S.F.-F.)
- Comprehensive Cancer Center Freiburg-CCCF, Medical Center-University of Freiburg, 79106 Freiburg, Germany
| | - Sven A. Lang
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.G.); (H.A.); (K.J.); (J.H.); (S.F.-F.)
- Department of Surgery and Transplantation, University Hospital RWTH, 52074 Aachen, Germany
- Correspondence:
| |
Collapse
|
19
|
Sugase T, Lam BQ, Danielson M, Terai M, Aplin AE, Gutkind JS, Sato T. Development and optimization of orthotopic liver metastasis xenograft mouse models in uveal melanoma. J Transl Med 2020; 18:208. [PMID: 32434572 PMCID: PMC7240939 DOI: 10.1186/s12967-020-02377-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/13/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with metastatic uveal melanoma (MUM) in the liver usually die within 1 year. The development of new treatments for MUM has been limited by the lack of diverse MUM cell lines and appropriate animal models. We previously reported that orthotopic xenograft mouse models established by direct injection of MUM cells into the liver were useful for the analysis associated with tumor microenvironment in the liver. However, considering that patients with UM metastasize to the liver hematogenously, direct liver injection model might not be suitable for investigation on various mechanisms of liver metastasis. Here, we aim to establish new orthotopic xenograft models via hematogenous dissemination of tumor cells to the liver, and to compare their characteristics with the hepatic injection model. We also determine if hepatic tumors could be effectively monitored with non-invasive live imaging. METHODS tdtTomate-labeled, patient-derived MUM cells were injected into the liver, spleen or tail vein of immunodeficient NSG mice. Tumor growth was serially assessed with In Vivo Imaging System (IVIS) images once every week. Established hepatic tumors were evaluated with CT scan and then analyzed histologically. RESULTS We found that splenic injection could consistently establish hepatic tumors. Non-invasive imaging showed that the splenic injection model had more consistent and stronger fluorescent intensity compared to the hepatic injection model. There were no significant differences in tumor growth between splenic injection with splenectomy and without splenectomy. The splenic injection established hepatic tumors diffusely throughout the liver, while the hepatic injection of tumor cells established a single localized tumor. Long-term monitoring of tumor development showed that tumor growth, tumor distribution in the liver, and overall survival depended on the number of tumor cells injected to the spleen. CONCLUSION We established a new orthotopic hepatic metastatic xenograft mouse model by splenic injection of MUM cells. The growth of orthotopic hepatic tumors could be monitored with non-invasive IVIS imaging. Moreover, we evaluated the therapeutic effect of a MEK inhibitor by using this model. Our findings suggest that our new orthotopic liver metastatic mouse model may be useful for preclinical drug screening experiments and for the analysis of liver metastasis mechanisms.
Collapse
Affiliation(s)
- Takahito Sugase
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1015 Walnut Street, Ste. 1024, Philadelphia, PA, USA
| | - Bao Q Lam
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1015 Walnut Street, Ste. 1024, Philadelphia, PA, USA
| | - Meggie Danielson
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1015 Walnut Street, Ste. 1024, Philadelphia, PA, USA
| | - Mizue Terai
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1015 Walnut Street, Ste. 1024, Philadelphia, PA, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - J Silvio Gutkind
- Department of Pharmacology, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Takami Sato
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1015 Walnut Street, Ste. 1024, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Lainas P, Rodda GA, Rafek G, Millereux M, Dammaro C, Trouiller P, Maitre S, Dagher I. Lifesaving transarterial embolization using absorbable gelatin sponge particles for massive bleeding of ruptured metastatic hepatic melanoma. Hepatobiliary Pancreat Dis Int 2020; 19:194-196. [PMID: 31822392 DOI: 10.1016/j.hbpd.2019.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 11/19/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Panagiotis Lainas
- Department of Minimally Invasive Digestive Surgery, Antoine-Beclere Hospital, AP-HP, Clamart F-92140, France; Paris-Saclay University, Orsay F-91405, France.
| | - Giorgia Amy Rodda
- Department of Minimally Invasive Digestive Surgery, Antoine-Beclere Hospital, AP-HP, Clamart F-92140, France
| | - George Rafek
- Department of Radiology, Antoine-Beclere Hospital, AP-HP, Clamart F-92140, France
| | - Maude Millereux
- Department of Intensive Care Unit, Antoine-Beclere Hospital, AP-HP, Clamart F-92140, France
| | - Carmelisa Dammaro
- Department of Minimally Invasive Digestive Surgery, Antoine-Beclere Hospital, AP-HP, Clamart F-92140, France; Paris-Saclay University, Orsay F-91405, France
| | - Pierre Trouiller
- Department of Intensive Care Unit, Antoine-Beclere Hospital, AP-HP, Clamart F-92140, France
| | - Sophie Maitre
- Department of Radiology, Antoine-Beclere Hospital, AP-HP, Clamart F-92140, France
| | - Ibrahim Dagher
- Department of Minimally Invasive Digestive Surgery, Antoine-Beclere Hospital, AP-HP, Clamart F-92140, France; Paris-Saclay University, Orsay F-91405, France
| |
Collapse
|
21
|
Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol 2020; 21:183-203. [PMID: 31937935 PMCID: PMC7102936 DOI: 10.1038/s41580-019-0199-y] [Citation(s) in RCA: 1618] [Impact Index Per Article: 323.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2019] [Indexed: 12/21/2022]
Abstract
The mTOR pathway integrates a diverse set of environmental cues, such as growth factor signals and nutritional status, to direct eukaryotic cell growth. Over the past two and a half decades, mapping of the mTOR signalling landscape has revealed that mTOR controls biomass accumulation and metabolism by modulating key cellular processes, including protein synthesis and autophagy. Given the pathway's central role in maintaining cellular and physiological homeostasis, dysregulation of mTOR signalling has been implicated in metabolic disorders, neurodegeneration, cancer and ageing. In this Review, we highlight recent advances in our understanding of the complex regulation of the mTOR pathway and discuss its function in the context of physiology, human disease and pharmacological intervention.
Collapse
Affiliation(s)
- Grace Y Liu
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute, Cambridge, MA, USA
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute, Cambridge, MA, USA.
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA.
| |
Collapse
|
22
|
Zheng B, Zhang S, Cai W, Wang J, Wang T, Tang N, Shi Y, Luo X, Yan W. Identification of Novel Fusion Transcripts in Undifferentiated Pleomorphic Sarcomas by Transcriptome Sequencing. Cancer Genomics Proteomics 2020; 16:399-408. [PMID: 31467233 DOI: 10.21873/cgp.20144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND/AIM Undifferentiated pleomorphic sarcoma (UPS) is an aggressive mesenchymal neoplasm characterized by chromosomal instability. The aim of this study was to identify fusion events involved in UPS. MATERIALS AND METHODS Transcriptome sequencing was performed to search for new fusion genes in 19 UPS samples, including two paired recurrent (R) and re-recurrent (RR) samples. RESULTS A total of 66 fusion genes were detected. Among them, 10 novel fusion genes were further confirmed by reverse transcription polymerase chain reaction (RT-PCR) and Sanger sequencing. Retinoblastoma (RB1) fusions (2 cases) were the most recurrent fusion genes. The gene fusions RB1-RNASEH2B, RB1-FGF14-AS1, and E2F6-FKBP4 were correlated with the Rb/E2F pathway. Pseudogenes were involved in the formation of the gene fusions CIC-DUX4L8 and EIF2AK4-ANXA2P2. Importantly, targetable gene fusions (PDGFRA-MACROD2 and NCOR1-MAP2K1) were detected in UPS. CONCLUSION Screening for the presence of fusion transcripts will provide vital clues to the understanding of genetic alterations and the finding of new targeted therapies for UPS.
Collapse
Affiliation(s)
- Biqiang Zheng
- Department of Musculoskeletal Cancer Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | | | - Weiluo Cai
- Department of Musculoskeletal Cancer Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Jian Wang
- Department of Musculoskeletal Cancer Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Ting Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Ning Tang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Yingqiang Shi
- Department of Musculoskeletal Cancer Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Xiaoying Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Wangjun Yan
- Department of Musculoskeletal Cancer Surgery, Fudan University Shanghai Cancer Center, Shanghai, P.R. China .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| |
Collapse
|
23
|
Krencz I, Sebestyen A, Papay J, Lou Y, Lutz GF, Majewicz TL, Khoor A. Correlation between immunohistochemistry and RICTOR fluorescence in situ hybridization amplification in small cell lung carcinoma. Hum Pathol 2019; 93:74-80. [PMID: 31454632 DOI: 10.1016/j.humpath.2019.08.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/16/2019] [Accepted: 08/18/2019] [Indexed: 12/23/2022]
Abstract
Small cell lung carcinoma (SCLC) accounts for approximately 15% of all lung cancers and remains a challenging disease, with no significant improvement in the field of targeted therapies. The RICTOR gene (rapamycin-insensitive companion of mTOR [mammalian target of rapamycin]), which encodes a key structural (scaffold) protein of mTOR complex 2), has recently been identified as one of the most frequently amplified genes and a potential therapeutic target in SCLC. The aim of this study was to compare immunohistochemical (IHC) expression of Rictor and phospho-Akt (a downstream target of mTOR complex 2) with RICTOR amplification as detected by fluorescence in situ hybridization (FISH) in SCLC. RICTOR FISH and Rictor and phospho-Akt IHC staining were performed on 100 formalin-fixed, paraffin-embedded SCLC samples. RICTOR amplification was detected in 15 samples (15%). IHC positivity for Rictor and phospho-Akt was observed in 37 (37%) and 42 (42%) samples, respectively. Considering FISH as the diagnostic standard, the sensitivity and specificity of Rictor IHC were 93% and 73%, whereas the sensitivity and specificity of phospho-Akt IHC were 80% and 65%, respectively. Rictor expression was higher in distant metastases than in primary tumor samples and lymph node metastases. There was no association between RICTOR amplification and clinical outcome. However, high expression of either Rictor or phospho-Akt was associated with significantly decreased overall survival. In conclusion, IHC expression of Rictor correlates highly with RICTOR amplification. Therefore, Rictor IHC can be used as a cost-effective method to select patients for RICTOR FISH and, potentially, for mTORC1/2 inhibitor therapy.
Collapse
Affiliation(s)
- Ildiko Krencz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary H-1085
| | - Anna Sebestyen
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary H-1085
| | - Judit Papay
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary H-1085
| | - Yanyan Lou
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL 32224
| | - Gabrielle F Lutz
- Clinical Research Internship Study Program, Mayo Clinic, Jacksonville, FL 32224
| | - Tracy L Majewicz
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Jacksonville, FL 32224
| | - Andras Khoor
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Jacksonville, FL 32224.
| |
Collapse
|
24
|
Chamcheu JC, Roy T, Uddin MB, Banang-Mbeumi S, Chamcheu RCN, Walker AL, Liu YY, Huang S. Role and Therapeutic Targeting of the PI3K/Akt/mTOR Signaling Pathway in Skin Cancer: A Review of Current Status and Future Trends on Natural and Synthetic Agents Therapy. Cells 2019; 8:cells8080803. [PMID: 31370278 PMCID: PMC6721560 DOI: 10.3390/cells8080803] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) and associated phosphatidyl-inositiol 3-kinase (PI3K)/protein kinase B (Akt) pathways regulate cell growth, differentiation, migration, and survival, as well as angiogenesis and metabolism. Dysregulation of these pathways is frequently associated with genetic/epigenetic alterations and predicts poor treatment outcomes in a variety of human cancers including cutaneous malignancies like melanoma and non-melanoma skin cancers. Recently, the enhanced understanding of the molecular and genetic basis of skin dysfunction in patients with skin cancers has provided a strong basis for the development of novel therapeutic strategies for these obdurate groups of skin cancers. This review summarizes recent advances in the roles of PI3K/Akt/mTOR and their targets in the development and progression of a broad spectrum of cutaneous cancers and discusses the current progress in preclinical and clinical studies for the development of PI3K/Akt/mTOR targeted therapies with nutraceuticals and synthetic small molecule inhibitors.
Collapse
Affiliation(s)
| | - Tithi Roy
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | | | - Sergette Banang-Mbeumi
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
- Division for Research and Innovation, POHOFI Inc., P.O. Box 44067, Madison, WI 53744, USA
- School of Nursing and Allied Health Sciences, Louisiana Delta Community College, Monroe, LA 71203, USA
| | | | - Anthony L Walker
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | - Yong-Yu Liu
- College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209-0497, USA
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| |
Collapse
|