1
|
Cokelaere C, Dok R, Cortesi EE, Zhao P, Sablina A, Nuyts S, Derua R, Janssens V. TIPRL1 and its ATM-dependent phosphorylation promote radiotherapy resistance in head and neck cancer. Cell Oncol (Dordr) 2024; 47:793-818. [PMID: 37971644 DOI: 10.1007/s13402-023-00895-6] [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] [Accepted: 10/23/2023] [Indexed: 11/19/2023] Open
Abstract
PURPOSE TIPRL1 (target of rapamycin signaling pathway regulator-like 1) is a known interactor and inhibitor of protein phosphatases PP2A, PP4 and PP6 - all pleiotropic modulators of the DNA Damage Response (DDR). Here, we investigated the role of TIPRL1 in the radiotherapy (RT) response of Head and Neck Squamous Cell Carcinoma (HNSCC). METHODS TIPRL1 mRNA (cBioportal) and protein expression (immunohistochemistry) in HNSCC samples were linked with clinical patient data. TIPRL1-depleted HNSCC cells were generated by CRISPR/Cas9 editing, and effects on colony growth, micronuclei formation (microscopy), cell cycle (flow cytometry), DDR signaling (immunoblots) and proteome (mass spectrometry) following RT were assessed. Mass spectrometry was used for TIPRL1 phosphorylation and interactomics analysis in irradiated cells. RESULTS TIPRL1 expression was increased in tumor versus non-tumor tissue, with high tumoral TIPRL1 expression associating with lower locoregional control and decreased survival of RT-treated patients. TIPRL1 deletion in HNSCC cells resulted in increased RT sensitivity, a faster but prolonged cell cycle arrest, increased micronuclei formation and an altered proteome-wide DDR. Upon irradiation, ATM phosphorylates TIPRL1 at Ser265. A non-phospho Ser265Ala mutant could not rescue the increased radiosensitivity phenotype of TIPRL1-depleted cells. While binding to PP2A-like phosphatases was confirmed, DNA-dependent protein kinase (DNA-PKcs), RAD51 recombinase and nucleosomal histones were identified as novel TIPRL1 interactors. Histone binding, although stimulated by RT, was adversely affected by TIPRL1 Ser265 phosphorylation. CONCLUSIONS Our findings underscore a clinically relevant role for TIPRL1 and its ATM-dependent phosphorylation in RT resistance through modulation of the DDR, highlighting its potential as a new HNSCC predictive marker and therapeutic target.
Collapse
Affiliation(s)
- Célie Cokelaere
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium
| | - Rüveyda Dok
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium
- Laboratory of Experimental Radiotherapy, Department of Oncology, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Emanuela E Cortesi
- Translational Cell & Tissue Research, Department of Imaging & Pathology, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Peihua Zhao
- VIB Laboratory of Mechanisms of Cell Transformation, Department of Oncology, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Anna Sablina
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium
- VIB Laboratory of Mechanisms of Cell Transformation, Department of Oncology, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Sandra Nuyts
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium
- Laboratory of Experimental Radiotherapy, Department of Oncology, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
- SybioMA, Proteomics Core Facility, University of Leuven (KU Leuven), B-3000, Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), B-3000, Leuven, Belgium.
- KU Leuven Cancer Institute (LKI), B-3000, Leuven, Belgium.
| |
Collapse
|
2
|
Domènech Omella J, Cortesi EE, Verbinnen I, Remmerie M, Wu H, Cubero FJ, Roskams T, Janssens V. A Novel Mouse Model of Combined Hepatocellular-Cholangiocarcinoma Induced by Diethylnitrosamine and Loss of Ppp2r5d. Cancers (Basel) 2023; 15:4193. [PMID: 37627221 PMCID: PMC10453342 DOI: 10.3390/cancers15164193] [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: 07/14/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Primary liver cancer (PLC) can be classified in hepatocellular (HCC), cholangiocarcinoma (CCA), and combined hepatocellular-cholangiocarcinoma (cHCC-CCA). The molecular mechanisms involved in PLC development and phenotype decision are still not well understood. Complete deletion of Ppp2r5d, encoding the B56δ subunit of Protein Phosphatase 2A (PP2A), results in spontaneous HCC development in mice via a c-MYC-dependent mechanism. In the present study, we aimed to examine the role of Ppp2r5d in an independent mouse model of diethylnitrosamine (DEN)-induced hepatocarcinogenesis. Ppp2r5d deletion (heterozygous and homozygous) accelerated HCC development, corroborating its tumor-suppressive function in liver and suggesting Ppp2r5d may be haploinsufficient. Ppp2r5d-deficient HCCs stained positively for c-MYC, consistent with increased AKT activation in pre-malignant and tumor tissues of Ppp2r5d-deficient mice. We also found increased YAP activation in Ppp2r5d-deficient tumors. Remarkably, in older mice, Ppp2r5d deletion resulted in cHCC-CCA development in this model, with the CCA component showing increased expression of progenitor markers (SOX9 and EpCAM). Finally, we observed an upregulation of Ppp2r5d in tumors from wildtype and heterozygous mice, revealing a tumor-specific control mechanism of Ppp2r5d expression, and suggestive of the involvement of Ppp2r5d in a negative feedback regulation restricting tumor growth. Our study highlights the tumor-suppressive role of mouse PP2A-B56δ in both HCC and cHCC-CCA, which may have important implications for human PLC development and targeted treatment.
Collapse
Affiliation(s)
- Judit Domènech Omella
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (J.D.O.); (I.V.); (M.R.)
| | - Emanuela E. Cortesi
- Translational Cell & Tissue Research, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (E.E.C.); (T.R.)
| | - Iris Verbinnen
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (J.D.O.); (I.V.); (M.R.)
| | - Michiel Remmerie
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (J.D.O.); (I.V.); (M.R.)
| | - Hanghang Wu
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; (H.W.); (F.J.C.)
| | - Francisco J. Cubero
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; (H.W.); (F.J.C.)
- Health Research Institute Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Centre for Biomedical Research, Network on Liver and Digestive Diseases (CIBEREHD), 28029 Madrid, Spain
| | - Tania Roskams
- Translational Cell & Tissue Research, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (E.E.C.); (T.R.)
- Department of Pathology, University Hospitals Leuven (UZ Leuven), 3000 Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Department of Cellular & Molecular Medicine, University of Leuven (KU Leuven), 3000 Leuven, Belgium; (J.D.O.); (I.V.); (M.R.)
- KU Leuven Cancer Institute (LKI), 3000 Leuven, Belgium
| |
Collapse
|
3
|
van Pelt J, Meeusen B, Derua R, Guffens L, Van Cutsem E, Janssens V, Verslype C. Human pancreatic cancer patients with Epithelial-to-Mesenchymal Transition and an aggressive phenotype show a disturbed balance in Protein Phosphatase Type 2A expression and functionality. J Transl Med 2023; 21:317. [PMID: 37170215 PMCID: PMC10176933 DOI: 10.1186/s12967-023-04145-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a low survival, its incidence is rising and little therapeutic improvements are expected in the near future. It has been observed that Epithelial-to-Mesenchymal transition (EMT) contributes (including in PDAC) to a more aggressive cancer phenotype. Additionally, largely unexplored, studies indicate a mechanistic interplay between Protein Phosphatase Type 2A (PP2A) enzymes and EMT that could offer treatment opportunities. The aim was to investigate the relation of a PP2A expression signature (encompassing all PP2A subunits, endogenous inhibitors and activators) with EMT and aggressive pancreatic cancer, and to discuss possible implications. METHODS We retrieved different PDAC expression datasets from NCBI to capture the variation in patients, and analyzed these using datamining, survival analysis, differential gene and protein expression. We determined genes highly associated with aggressive PDAC. For in vitro evaluation, Panc-1 cells were treated with the pharmacologic PP2A inhibitor Okadaic Acid (OA). Additionally, two OA-resistant Panc-1 clones were developed and characterized. RESULTS In patients, there is a strong correlation between EMT and aggressive PDAC, and between aggressive PDAC and PP2A, with a significant upregulation of PP2A inhibitor genes. Several PP2A genes significantly correlated with decreased survival. In vitro, short-term exposure to OA induced EMT in Panc-1 cells. This shift towards EMT was further pronounced in the OA-resistant Panc-1 clones, morphologically and by pathway analysis. Proteomic analysis and gene sequencing showed that the advanced OA-resistant model most resembles the clinical PDAC presentation (with EMT signature, and with several specific PP2A genes upregulated, and others downregulated). CONCLUSIONS We demonstrated a strong association between EMT, altered PP2A expression and aggressive PDAC in patients. Also, in vitro, PP2A inhibition induces EMT. Overall, statistics suggests the mechanistic importance of PP2A dysregulation for PDAC progression. Translationally, our observations indicate that pharmacologic restoration of PP2A activity could be an attractive therapeutic strategy to block or reverse progression.
Collapse
Affiliation(s)
- Jos van Pelt
- Laboratory of Digestive Oncology, Department of Oncology, KU Leuven & University Hospitals Leuven, Geb. Onderwijs & Navorsing 4, Room 07.465, Herestraat 49, Bus 603, B3000, Leuven, Belgium.
- KU Leuven Cancer Institute (LKI), Herestraat 49, B3000, Leuven, Belgium.
| | - Bob Meeusen
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, O&N1, University of Leuven (KU Leuven), Herestraat 49, Bus 901, B3000, Leuven, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, O&N1, University of Leuven (KU Leuven), Herestraat 49, Bus 901, B3000, Leuven, Belgium
- SyBioMa (KU Leuven), Herestraat 49, B3000, Leuven, Belgium
| | - Liesbeth Guffens
- KU Leuven Cancer Institute (LKI), Herestraat 49, B3000, Leuven, Belgium
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, O&N1, University of Leuven (KU Leuven), Herestraat 49, Bus 901, B3000, Leuven, Belgium
| | - Eric Van Cutsem
- Laboratory of Digestive Oncology, Department of Oncology, KU Leuven & University Hospitals Leuven, Geb. Onderwijs & Navorsing 4, Room 07.465, Herestraat 49, Bus 603, B3000, Leuven, Belgium
- KU Leuven Cancer Institute (LKI), Herestraat 49, B3000, Leuven, Belgium
| | - Veerle Janssens
- KU Leuven Cancer Institute (LKI), Herestraat 49, B3000, Leuven, Belgium.
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, O&N1, University of Leuven (KU Leuven), Herestraat 49, Bus 901, B3000, Leuven, Belgium.
| | - Chris Verslype
- Laboratory of Digestive Oncology, Department of Oncology, KU Leuven & University Hospitals Leuven, Geb. Onderwijs & Navorsing 4, Room 07.465, Herestraat 49, Bus 603, B3000, Leuven, Belgium
- KU Leuven Cancer Institute (LKI), Herestraat 49, B3000, Leuven, Belgium
| |
Collapse
|
4
|
Arribas RL, Viejo L, Bravo I, Martínez M, Ramos E, Romero A, García-Frutos EM, Janssens V, Montiel C, de Los Ríos C. C-glycosides analogues of the okadaic acid central fragment exert neuroprotection via restoration of PP2A-phosphatase activity: A rational design of potential drugs for Alzheimer's disease targeting tauopathies. Eur J Med Chem 2023; 251:115245. [PMID: 36905916 DOI: 10.1016/j.ejmech.2023.115245] [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: 10/31/2022] [Revised: 01/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Protein phosphatase 2A (PP2A) is an important Ser/Thr phosphatase that participates in the regulation of multiple cellular processes. This implies that any deficient activity of PP2A is the responsible of severe pathologies. For instance, one of the main histopathological features of Alzheimer's disease is neurofibrillary tangles, which are mainly comprised by hyperphosphorylated forms of tau protein. This altered rate of tau phosphorylation has been correlated with PP2A depression AD patients. With the goal of preventing PP2A inactivation in neurodegeneration scenarios, we have aimed to design, synthesize and evaluate new ligands of PP2A capable of preventing its inhibition. To achieve this goal, the new PP2A ligands present structural similarities with the central fragment C19-C27 of the well-established PP2A inhibitor okadaic acid (OA). Indeed, this central moiety of OA does not exert inhibitory actions. Hence, these compounds lack PP2A-inhibiting structural motifs but, in contrast, compete with PP2A inhibitors, thus recovering phosphatase activity. Proving this hypothesis, most compounds showed a good neuroprotective profile in neurodegeneration models related to PP2A impairment, highlighting derivative 10, named ITH12711, as the most promising one. This compound (1) restored in vitro and cellular PP2A catalytic activity, measured on a phospho-peptide substrate and by western-blot analyses, (2) proved good brain penetration measured by PAMPA, and (3) prevented LPS-induced memory impairment of mice in the object recognition test. Thus, the promising outcomes of the compound 10 validate our rational approach to design new PP2A-activating drugs based on OA central fragment.
Collapse
Affiliation(s)
- Raquel L Arribas
- Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Spain
| | - Lucía Viejo
- Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/ Diego de León, 62, 28006, Madrid, Spain
| | - Isaac Bravo
- Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/ Diego de León, 62, 28006, Madrid, Spain; Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, 28049, Madrid, Spain
| | - Minerva Martínez
- Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | - Eva Ramos
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense, 28040, Madrid, Spain
| | - Alejandro Romero
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense, 28040, Madrid, Spain
| | - Eva M García-Frutos
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, 28049, Madrid, Spain; Universidad de Alcalá, Departamento de Química Orgánica y Química Inorgánica, Ctra. Madrid-Barcelona Km.33,600, 28871, Alcalá de Henares, Madrid, Spain
| | - Veerle Janssens
- Department of Cellular & Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, B-3000, Leuven, Belgium; LBI (KU Leuven Brain Institute), B-3000, Leuven, Belgium
| | - Carmen Montiel
- Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | - Cristóbal de Los Ríos
- Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, 28029, Madrid, Spain; Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Spain; Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, C/ Diego de León, 62, 28006, Madrid, Spain.
| |
Collapse
|
5
|
Baietti MF, Sewduth RN. Novel Therapeutic Approaches Targeting Post-Translational Modifications in Lung Cancer. Pharmaceutics 2023; 15:pharmaceutics15010206. [PMID: 36678835 PMCID: PMC9865455 DOI: 10.3390/pharmaceutics15010206] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Lung cancer is one of the most common cancers worldwide. It consists of two different subtypes: non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). Despite novel therapeutic options such as immunotherapy, only 20% of lung cancer patients survive the disease after five years. This low survival rate is due to acquired drug resistance and severe off-target effects caused by currently used therapies. Identification and development of novel and targeted therapeutic approaches are urgently required to improve the standard of care for lung cancer patients. Here, we describe the recent development of novel drug-delivery approaches, such as adenovirus, lipid nanoparticles, and PROTACs, that have been tested in clinical trials and experimentally in the context of fundamental research. These different options show that it is now possible to target protein kinases, phosphatases, ubiquitin ligases, or protein modifications directly in lung cancer to block disease progression. Furthermore, the recent acceptance of RNA vaccines using lipid nanoparticles has further revealed therapeutic options that could be combined with chemo-/immunotherapies to improve current lung cancer therapies. This review aims to compare recent advances in the pharmaceutical research field for the development of technologies targeting post-translational modifications or protein modifiers involved in the tumorigenesis of lung cancer.
Collapse
Affiliation(s)
- Maria Francesca Baietti
- TRACE, Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Correspondence: (M.F.B.); (R.N.S.)
| | - Raj Nayan Sewduth
- VIB-KU Leuven Center for Cancer Biology, Herestraat 49, 3000 Leuven, Belgium
- Correspondence: (M.F.B.); (R.N.S.)
| |
Collapse
|
6
|
Haanen TJ, O'Connor CM, Narla G. Biased holoenzyme assembly of protein phosphatase 2A (PP2A): From cancer to small molecules. J Biol Chem 2022; 298:102656. [PMID: 36328247 PMCID: PMC9707111 DOI: 10.1016/j.jbc.2022.102656] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a family of serine threonine phosphatases responsible for regulating protein phosphorylation, thus opposing the activity of cellular kinases. PP2A is composed of a catalytic subunit (PP2A Cα/β) and scaffolding subunit (PP2A Aα/β) and various substrate-directing B regulatory subunits. PP2A biogenesis is regulated at multiple levels. For example, the sequestration of the free catalytic subunit during the process of biogenesis avoids promiscuous phosphatase activity. Posttranslational modifications of PP2A C direct PP2A heterotrimeric formation. Additionally, PP2A functions as a haploinsufficient tumor suppressor, where attenuated PP2A enzymatic activity creates a permissive environment for oncogenic transformation. Recent work studying PP2A in cancer showed that its role in tumorigenesis is more nuanced, with some holoenzymes being tumor suppressive, while others are required for oncogenic transformation. In cancer biology, PP2A function is modulated through various mechanisms including the displacement of specific B regulatory subunits by DNA tumor viral antigens, by recurrent mutations, and through loss of carboxymethyl-sensitive heterotrimeric complexes. In aggregate, these alterations bias PP2A activity away from its tumor suppressive functions and toward oncogenic ones. From a therapeutic perspective, molecular glues and disruptors present opportunities for both the selective stabilization of tumor-suppressive holoenzymes and disruption of holoenzymes that are pro-oncogenic. Collectively, these approaches represent an attractive cancer therapy for a wide range of tumor types. This review will discuss the mechanisms by which PP2A holoenzyme formation is dysregulated in cancer and the current therapies that are aimed at biasing heterotrimer formation of PP2A for the treatment of cancer.
Collapse
Affiliation(s)
- Terrance J Haanen
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA
| | - Caitlin M O'Connor
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
7
|
Nussinov R, Tsai CJ, Jang H. A New View of Activating Mutations in Cancer. Cancer Res 2022; 82:4114-4123. [PMID: 36069825 PMCID: PMC9664134 DOI: 10.1158/0008-5472.can-22-2125] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/16/2022] [Accepted: 09/01/2022] [Indexed: 12/14/2022]
Abstract
A vast effort has been invested in the identification of driver mutations of cancer. However, recent studies and observations call into question whether the activating mutations or the signal strength are the major determinant of tumor development. The data argue that signal strength determines cell fate, not the mutation that initiated it. In addition to activating mutations, factors that can impact signaling strength include (i) homeostatic mechanisms that can block or enhance the signal, (ii) the types and locations of additional mutations, and (iii) the expression levels of specific isoforms of genes and regulators of proteins in the pathway. Because signal levels are largely decided by chromatin structure, they vary across cell types, states, and time windows. A strong activating mutation can be restricted by low expression, whereas a weaker mutation can be strengthened by high expression. Strong signals can be associated with cell proliferation, but too strong a signal may result in oncogene-induced senescence. Beyond cancer, moderate signal strength in embryonic neural cells may be associated with neurodevelopmental disorders, and moderate signals in aging may be associated with neurodegenerative diseases, like Alzheimer's disease. The challenge for improving patient outcomes therefore lies in determining signaling thresholds and predicting signal strength.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, NCI, Frederick, Maryland
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, NCI, Frederick, Maryland
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, NCI, Frederick, Maryland
| |
Collapse
|
8
|
Selumetinib: a selective MEK1 inhibitor for solid tumor treatment. Clin Exp Med 2022; 23:229-244. [PMID: 35171389 DOI: 10.1007/s10238-021-00783-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022]
Abstract
Cancer incidence is rapidly growing. Solid tumors are responsible for a majority of cancers. Recently, molecular-targeted agents have played a significant role in cancer treatment. Ras-Raf-MEK-ERK signaling pathway, is a substantial element in the survival, propagation, and drug resistance of human cancers. MEK is a specific part of the so-called cascade, and ERK proteins are its sole target. Furthermore, their downstream position in the Ras-ERK cascade, is noteworthy to direct their function in patients with upstream mutated genes. MEK1 mutations are responsible for initiating several solid tumors. Selumetinib (AZD6244) is a second-generation, selective, potent, and non-ATP competitive allosteric MEK1 inhibitor. The efficacy of selumetinib in various solid tumors such as colorectal cancer, lung cancer, neurofibroma, and melanoma is investigated. The present paper provides an overview of the MAPK cascade, the role of selumetinib as a MEK1/2 inhibitor, and the related findings of clinical trials for solid tumor treatment.
Collapse
|