14 results on '"Grossi, Valentina"'
Search Results
2. Clinical Assessment and Genetic Testing for Hereditary Polyposis Syndromes in an Italian Cohort of Patients with Colorectal Polyps.
- Author
-
Fasano, Candida, Cariola, Filomena, Forte, Giovanna, Buonadonna, Antonia Lucia, Sanese, Paola, Manghisi, Andrea, Lepore Signorile, Martina, De Marco, Katia, Grossi, Valentina, Disciglio, Vittoria, and Simone, Cristiano
- Subjects
RISK assessment ,RESEARCH funding ,COLORECTAL cancer ,INTESTINAL polyps ,COLON polyps ,LONGITUDINAL method ,GENETIC variation ,GENETIC disorders ,GENE expression profiling ,GENETIC mutation ,GENETIC testing ,HEREDITARY cancer syndromes ,SEQUENCE analysis ,PHENOTYPES ,DISEASE risk factors - Abstract
Simple Summary: This study reports the results of genetic testing to identify germline variants in the main genes (APC, BMPR1A, MUTYH, PTEN, SMAD4, STK11) associated with hereditary polyposis syndromes in 75 index cases with colorectal polyps and a personal/family history of cancer that had been referred to genetic counseling at the Medical Genetics Unit of the National Institute of Gastroenterology "Saverio de Bellis", Castellana Grotte, Bari, Italy. In the screened patients, some of which did not meet the recommended eligibility criteria of current National Comprehensive Cancer Network (NCCN) guidelines for genetic testing, we identified 14 pathogenic variants and 6 variants of uncertain significance. Of note, by combining the results of multigene panel tests with the evaluation of patients' clinical phenotype and family history, we were able to confirm the diagnosis of hereditary polyposis syndrome for pathogenic variant carriers and assign them to specific clinical surveillance and management programs. Background: Hereditary polyposis syndromes are clinically and genetically heterogeneous conditions associated with increased colorectal cancer risk. They are classified based on polyp histology, inheritance mode, causal gene, and colonic and extracolonic manifestations. Their diagnosis is challenging due to overlapping and heterogeneous clinical presentations. Methods: A multigene next-generation sequencing panel was used to screen 75 index cases with colorectal polyps and a personal/family history of cancer for key hereditary polyposis-associated genes (APC, BMPR1A, MUTYH, PTEN, SMAD4, and STK11) in order to identify germline genetic variants. Results: In the screened index cases, we found 14 pathogenic variants involving APC, MUTYH, SMAD4, and STK11 and 6 variants of uncertain significance involving APC, BMPR1A, and SMAD4. In this cohort, four patients not fulfilling the recommended eligibility criteria of current National Comprehensive Cancer Network (NCCN) guidelines for genetic testing were molecularly diagnosed with a hereditary polyposis syndrome. Conclusions: Our findings indicate that stringent NCCN eligibility criteria for molecular screening may lead to missing some of the patients affected by hereditary polyposis syndromes. This highlights the need for a careful evaluation of patients' clinical manifestations, polyp number, age of polyp onset, and family history to select appropriate candidates for molecular diagnosis of these conditions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Tumor Testing and Genetic Analysis to Identify Lynch Syndrome Patients in an Italian Colorectal Cancer Cohort.
- Author
-
Pantaleo, Antonino, Forte, Giovanna, Cariola, Filomena, Valentini, Anna Maria, Fasano, Candida, Sanese, Paola, Grossi, Valentina, Buonadonna, Antonia Lucia, De Marco, Katia, Lepore Signorile, Martina, Guglielmi, Anna Filomena, Manghisi, Andrea, Gigante, Gianluigi, Armentano, Raffaele, Disciglio, Vittoria, and Simone, Cristiano
- Subjects
DIAGNOSIS of hereditary nonpolyposis colorectal cancer ,EARLY detection of cancer ,HEREDITARY nonpolyposis colorectal cancer ,COLORECTAL cancer ,DNA methylation ,RESEARCH funding ,DESCRIPTIVE statistics ,LONGITUDINAL method - Abstract
Simple Summary: Lynch syndrome (LS) is an inherited genetic condition caused by germline mutations in DNA mismatch repair (MMR) genes. It is associated with a predisposition to different types of cancer, including colorectal cancer (CRC). CRC is the fourth most common cancer worldwide. The screening algorithm for the selection of LS patients is based on the identification of CRC specimens that have MMR loss/high microsatellite instability (MSI-H) and are wild-type for BRAF
V600 . The aim of this retrospective study was to clinically and molecularly characterize CRC patients with these features. We used a comprehensive approach including tumor testing for the assessment of MSI status, clinical evaluation of patients and their families, and genetic analysis to identify variants in MMR and other cancer-related genes. The clinical and molecular characterization of these patients highlights the importance of personalized medicine to provide tailored genetic counseling, management, and surveillance to families with LS and hereditary cancer. Lynch syndrome (LS) is an inherited cancer susceptibility syndrome caused by germline mutations in a DNA mismatch repair (MMR) gene or in the EPCAM gene. LS is associated with an increased lifetime risk of colorectal cancer (CRC) and other malignancies. The screening algorithm for LS patient selection is based on the identification of CRC specimens that have MMR loss/high microsatellite instability (MSI-H) and are wild-type for BRAFV600 . Here, we sought to clinically and molecularly characterize patients with these features. From 2017 to 2023, 841 CRC patients were evaluated for MSI and BRAFV600E mutation status, 100 of which showed MSI-H. Of these, 70 were wild-type for BRAFV600 . Among these 70 patients, 30 were genetically tested for germline variants in hereditary cancer predisposition syndrome genes. This analysis showed that 19 of these 30 patients (63.3%) harbored a germline pathogenic or likely pathogenic variant in MMR genes, 2 (6.7%) harbored a variant of unknown significance (VUS) in MMR genes, 3 (10%) harbored a VUS in other cancer-related genes, and 6 (20%) were negative to genetic testing. These findings highlight the importance of personalized medicine for tailored genetic counseling, management, and surveillance of families with LS and other hereditary cancer syndromes. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
4. Correction: Lepore Signorile et al. c-MYC Protein Stability Is Sustained by MAPKs in Colorectal Cancer. Cancers 2022, 14 , 4840.
- Author
-
Lepore Signorile, Martina, Grossi, Valentina, Fasano, Candida, Forte, Giovanna, Disciglio, Vittoria, Sanese, Paola, De Marco, Katia, La Rocca, Francesca, Armentano, Raffaele, Valentini, Anna Maria, Giannelli, Gianluigi, and Simone, Cristiano
- Subjects
- *
MITOGEN-activated protein kinases , *COLORECTAL cancer , *ONCOGENES - Published
- 2024
- Full Text
- View/download PDF
5. Colorectal Cancer Chemoprevention: A Dream Coming True?
- Author
-
Lepore Signorile, Martina, Grossi, Valentina, Fasano, Candida, and Simone, Cristiano
- Subjects
- *
CANCER chemoprevention , *COLORECTAL cancer , *FECAL occult blood tests , *PRECANCEROUS conditions - Abstract
Colorectal cancer (CRC) is one of the deadliest forms of cancer worldwide. CRC development occurs mainly through the adenoma-carcinoma sequence, which can last decades, giving the opportunity for primary prevention and early detection. CRC prevention involves different approaches, ranging from fecal occult blood testing and colonoscopy screening to chemoprevention. In this review, we discuss the main findings gathered in the field of CRC chemoprevention, focusing on different target populations and on various precancerous lesions that can be used as efficacy evaluation endpoints for chemoprevention. The ideal chemopreventive agent should be well tolerated and easy to administer, with low side effects. Moreover, it should be readily available at a low cost. These properties are crucial because these compounds are meant to be used for a long time in populations with different CRC risk profiles. Several agents have been investigated so far, some of which are currently used in clinical practice. However, further investigation is needed to devise a comprehensive and effective chemoprevention strategy for CRC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Short Linear Motifs in Colorectal Cancer Interactome and Tumorigenesis.
- Author
-
Fasano, Candida, Grossi, Valentina, Forte, Giovanna, and Simone, Cristiano
- Subjects
- *
COLORECTAL cancer , *SMALL molecules , *NEOPLASTIC cell transformation , *PROTEIN domains , *CELL communication - Abstract
Colorectal tumorigenesis is driven by alterations in genes and proteins responsible for cancer initiation, progression, and invasion. This multistage process is based on a dense network of protein–protein interactions (PPIs) that become dysregulated as a result of changes in various cell signaling effectors. PPIs in signaling and regulatory networks are known to be mediated by short linear motifs (SLiMs), which are conserved contiguous regions of 3–10 amino acids within interacting protein domains. SLiMs are the minimum sequences required for modulating cellular PPI networks. Thus, several in silico approaches have been developed to predict and analyze SLiM-mediated PPIs. In this review, we focus on emerging evidence supporting a crucial role for SLiMs in driver pathways that are disrupted in colorectal cancer (CRC) tumorigenesis and related PPI network alterations. As a result, SLiMs, along with short peptides, are attracting the interest of researchers to devise small molecules amenable to be used as novel anti-CRC targeted therapies. Overall, the characterization of SLiMs mediating crucial PPIs in CRC may foster the development of more specific combined pharmacological approaches. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. c-MYC Protein Stability Is Sustained by MAPKs in Colorectal Cancer.
- Author
-
Lepore Signorile, Martina, Grossi, Valentina, Fasano, Candida, Forte, Giovanna, Disciglio, Vittoria, Sanese, Paola, De Marco, Katia, La Rocca, Francesca, Armentano, Raffaele, Valentini, Anna Maria, Giannelli, Gianluigi, and Simone, Cristiano
- Subjects
- *
IN vitro studies , *IN vivo studies , *ONCOGENES , *ANIMAL experimentation , *COLORECTAL cancer , *MITOGEN-activated protein kinases , *MICE - Abstract
Simple Summary: Colorectal cancer (CRC) is the most common gastrointestinal tract malignancy. Previous reports have shown that cancerous phenotypes in the intestine are dependent on c-MYC target gene expression. Unfortunately, finding c-MYC inhibitors has proven difficult because c-MYC does not have a deep surface-binding pocket. Considering that c-MYC is maintained upregulated through β-catenin-mediated transcriptional activation and ERK-mediated post-translational stabilization, and since we have previously demonstrated that c-MYC transcriptional activation is affected by p38α as a β-catenin chromatin-associated kinase, here, we investigated p38α's involvement in c-MYC protein stabilization in CRC. Interestingly, we found that p38α sustains c-MYC's stability by preventing its ubiquitination and proteasomal degradation. Moreover, we showed that p38α inhibitors exhibit a synthetic lethality effect when used in combination with MEK inhibitors in CRC cells. Our findings identify p38α as a promising therapeutic target that acts on the pharmacologically "undruggable" c-MYC protein, with implications for countering c-MYC-mediated CRC proliferation, metastasization, and chemoresistance. c-MYC is one of the most important factors involved in colorectal cancer (CRC) initiation and progression; indeed, it is found to be upregulated in up to 80% of sporadic cases. During colorectal carcinogenesis, c-MYC is maintained upregulated through β-catenin-mediated transcriptional activation and ERK-mediated post-translational stabilization. Our data demonstrate that p38α, a kinase involved in CRC metabolism and survival, contributes to c-Myc protein stability. Moreover, we show that p38α, like ERK, stabilizes c-MYC protein levels by preventing its ubiquitination. Of note, we found that p38α phosphorylates c-MYC and interacts with it both in vitro and in cellulo. Extensive molecular analyses in the cellular and in vivo models revealed that the p38α kinase inhibitors, SB202190 and ralimetinib, affect c-MYC protein levels. Ralimetinib also exhibited a synthetic lethality effect when used in combination with the MEK1 inhibitor trametinib. Overall, our findings identify p38α as a promising therapeutic target, acting directly on c-MYC, with potential implications for countering c-MYC-mediated CRC proliferation, metastatic dissemination, and chemoresistance. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Sorafenib inhibits p38α activity in colorectal cancer cells and synergizes with the DFG-in inhibitor SB202190 to increase apoptotic response.
- Author
-
Grossi, Valentina, Liuzzi, Micaela, Murzilli, Stefania, Martelli, Nicola, Napoli, Anna, Ingravallo, Giuseppe, Del Rio, Alberto, and Simone, Cristiano
- Published
- 2012
- Full Text
- View/download PDF
9. Blocking p38/ERK crosstalk affects colorectal cancer growth by inducing apoptosis in vitro and in preclinical mouse models
- Author
-
Chiacchiera, Fulvio, Grossi, Valentina, Cappellari, Marianna, Peserico, Alessia, Simonatto, Marta, Germani, Aldo, Russo, Silvana, Moyer, Mary P., Resta, Nicoletta, Murzilli, Stefania, and Simone, Cristiano
- Subjects
- *
COLON cancer , *TUMOR growth , *APOPTOSIS , *LABORATORY mice , *AUTOPHAGY , *CELL death , *CASPASES - Abstract
Abstract: We recently demonstrated that p38α is required to maintain colorectal cancer (CRC) metabolism, as its inhibition leads to FoxO3A activation, autophagy, cell death, and tumor growth reduction both in vitro and in vivo. Here we show that inhibition of p38α is followed by TRAIL-mediated activation of caspase-8 and FoxO3A-dependent HER3 upregulation with consequent overactivation of the MEK-ERK1/2 survival pathway. p38α and MEK combined inhibition specifically induces apoptosis by enabling TRAIL signaling propagation through t-Bid and caspase-3, and fosters cell death in CRC cells and preclinical mouse models. Current MEK1-directed pharmacological strategies could thus be exploited, in combination with p38α inhibition, to develop new approaches for CRC treatment. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
10. Updates from the Intestinal Front Line: Autophagic Weapons against Inflammation and Cancer.
- Author
-
Madia, Federica, Grossi, Valentina, Peserico, Alessia, and Simone, Cristiano
- Abstract
The intestine lies at the interface between the organism and its environment and responds to infection/inflammation in a multi-leveled manner, potentially leading to chronic inflammatory pathologies and cancer formation. Indeed, the immune response at the intestinal epithelium has been found to be involved in the origin and development of colorectal cancer, which is the third most commonly diagnosed neoplastic disease. Among the mechanisms induced upon inflammation, autophagy appears as a defensive strategy for the clearance of invading microbes and intracellular waste components. Autophagy has also been found to play an important role in colorectal cancer, where it seems to have a pro-survival or pro-death function depending on the stage of the neoplastic process. In this paper we discuss the dual role of autophagy in colorectal cancer and review evidence showing that modulation of autophagy affects the immune response and cancer biology. The study of key players involved in autophagy might contribute to the design of new approaches for colorectal cancer, consisting in combined therapies capable of modifying cancer-specific metabolism rather than simply evoking a generic apoptotic and/or autophagic response, thus enhancing the efficacy of currently used drugs and treatments. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
11. Chasing the FOXO3: Insights into Its New Mitochondrial Lair in Colorectal Cancer Landscape.
- Author
-
Grossi, Valentina, Fasano, Candida, Celestini, Valentina, Lepore Signorile, Martina, Sanese, Paola, and Simone, Cristiano
- Subjects
- *
CELLULAR signal transduction , *COLON tumors , *DRUG resistance in cancer cells , *HOMEOSTASIS , *MITOCHONDRIA , *PHOSPHORYLATION , *PHYSIOLOGICAL stress , *TRANSCRIPTION factors ,RECTUM tumors - Abstract
Colorectal cancer (CRC) poses a formidable challenge in terms of molecular heterogeneity, as it involves a variety of cancer-related pathways and molecular changes unique to an individual's tumor. On the other hand, recent advances in DNA sequencing technologies provide an unprecedented capacity to comprehensively identify the genetic alterations resulting in tumorigenesis, raising the hope that new therapeutic approaches based on molecularly targeted drugs may prevent the occurrence of chemoresistance. Regulation of the transcription factor FOXO3a in response to extracellular cues plays a fundamental role in cellular homeostasis, being part of the molecular machinery that drives cells towards survival or death. Indeed, FOXO3a is controlled by a range of external stimuli, which not only influence its transcriptional activity, but also affect its subcellular localization. These regulation mechanisms are mediated by cancer-related signaling pathways that eventually drive changes in FOXO3a post-translational modifications (e.g., phosphorylation). Recent results showed that FOXO3a is imported into the mitochondria in tumor cells and tissues subjected to metabolic stress and cancer therapeutics, where it induces expression of the mitochondrial genome to support mitochondrial metabolism and cell survival. The current review discusses the potential clinical relevance of multidrug therapies that drive cancer cell fate by regulating critical pathways converging on FOXO3a. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
12. Targeted therapy against chemoresistant colorectal cancers: Inhibition of p38α modulates the effect of cisplatin in vitro and in vivo through the tumor suppressor FoxO3A.
- Author
-
Germani, Aldo, Matrone, Antonio, Grossi, Valentina, Peserico, Alessia, Sanese, Paola, Liuzzi, Micaela, Palermo, Rocco, Murzilli, Stefania, Campese, Antonio Francesco, Ingravallo, Giuseppe, Canettieri, Gianluca, Tezil, Tugsan, and Simone, Cristiano
- Subjects
- *
COLON cancer treatment , *TARGETED drug delivery , *CISPLATIN , *IN vitro studies , *TUMOR suppressor genes , *MITOGEN-activated protein kinases , *CANCER chemotherapy - Abstract
Abstract: Chemoresistance is a major obstacle to effective therapy against colorectal cancer (CRC) and may lead to deadly consequences. The metabolism of CRC cells depends highly on the p38 MAPK pathway, whose involvement in maintaining a chemoresistant behavior is currently being investigated. Our previous studies revealed that p38α is the main p38 isoform in CRC cells. Here we show that p38α pharmacological inhibition combined with cisplatin administration decreases colony formation and viability of cancer cells and strongly increases Bax-dependent apoptotic cell death by activating the tumor suppressor protein FoxO3A. Our results indicate that FoxO3A activation up-regulates transcription of its target genes (p21, PTEN, Bim and GADD45), which forces both chemosensitive and chemoresistant CRC cells to undergo apoptosis. Additionally, we found that FoxO3A is required for apoptotic cell death induction, as confirmed by RNA interference experiments. In animal models xenografted with chemoresistant HT29 cells, we further confirmed that the p38-targeted dual therapy strategy produced an increase in apoptosis in cancer tissue leading to tumor regression. Our study uncovers a major role for the p38-FoxO3A axis in chemoresistance, thereby suggesting a new therapeutic approach for CRC treatment; moreover, our results indicate that Bax status may be used as a predictive biomarker. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
13. Discovery of the 4-aminopiperidine-based compound EM127 for the site-specific covalent inhibition of SMYD3.
- Author
-
Parenti, Marco Daniele, Naldi, Marina, Manoni, Elisabetta, Fabini, Edoardo, Cederfelt, Daniela, Talibov, Vladimir O., Gressani, Valeria, Guven, Ummu, Grossi, Valentina, Fasano, Candida, Sanese, Paola, De Marco, Katia, Shtil, Alexander A., Kurkin, Alexander V., Altieri, Andrea, Danielson, U. Helena, Caretti, Giuseppina, Simone, Cristiano, Varchi, Greta, and Bartolini, Manuela
- Subjects
- *
CANCER cells , *MASS spectrometry , *CELL lines , *METHYLTRANSFERASES , *COLORECTAL cancer , *CYCLIN-dependent kinases - Abstract
Recent findings support the hypothesis that inhibition of SMYD3 methyltransferase may be a therapeutic avenue for some of the deadliest cancer types. Herein, active site-selective covalent SMYD3 inhibitors were designed by introducing an appropriate reactive cysteine trap into reversible first-generation SMYD3 inhibitors. The 4-aminopiperidine derivative EM127 (11C) bearing a 2-chloroethanoyl group as reactive warhead showed selectivity for Cys186, located in the substrate/histone binding pocket. Selectivity towards Cys186 was retained even at high inhibitor/enzyme ratio, as shown by mass spectrometry. The mode of interaction with the SMYD3 substrate/histone binding pocket was revealed by crystallographic studies. In enzymatic assays, 11C showed a stronger SMYD3 inhibitory effect compared to the reference inhibitor EPZ031686. Remarkably, 11C attenuated the proliferation of MDA-MB-231 breast cancer cell line at the same low micromolar range of concentrations that reduced SMYD3 mediated ERK signaling in HCT116 colorectal cancer and MDA-MB-231 breast cancer cells. Furthermore, 11C (5 μM) strongly decreased the steady-state mRNA levels of genes important for tumor biology such as cyclin dependent kinase 2, c-MET, N-cadherin and fibronectin 1, all known to be regulated, at least in part, by SMYD3. Thus, 11C is as a first example of second generation SMYD3 inhibitors; this agent represents a covalent and a site specific SMYD3 binder capable of potent and prolonged attenuation of methyltransferase activity. [Display omitted] • The 4-aminopiperidine-based compound EM127 irreversibly inhibits SMYD3. • EM127 shows a striking selectivity towards the target Cys186 residue. • EM127 is more potent than the reference inhibitor EPZ031686. • X-ray structure of SMYD3-EM127 confirms the formation of a covalent adduct. • Cancer cell lines are highly sensitive to EM127 pharmacological inhibition. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
14. Pharmacological targeting of the novel β-catenin chromatin-associated kinase p38α in colorectal cancer stem cell tumorspheres and organoids
- Author
-
Gianluca Canettieri, Katia De Marco, Martina Lepore Signorile, Cristiano Simone, Gabriella Di Carlo, Valentina Grossi, Francesco Dituri, Giuseppe Ingravallo, Candida Fasano, Annalisa Nicotra, Giovanna Forte, Paola Sanese, Gianluigi Giannelli, Simone Di Franco, Giorgio Stassi, Francesco Susca, Laura Rosa Mangiapane, Vittoria Disciglio, Lepore Signorile, Martina, Grossi, Valentina, Di Franco, Simone, Forte, Giovanna, Disciglio, Vittoria, Fasano, Candida, Sanese, Paola, De Marco, Katia, Susca, Francesco Claudio, Mangiapane, Laura Rosa, Nicotra, Annalisa, Di Carlo, Gabriella, Dituri, Francesco, Giannelli, Gianluigi, Ingravallo, Giuseppe, Canettieri, Gianluca, Stassi, Giorgio, and Simone, Cristiano
- Subjects
Cancer Research ,Settore MED/06 - Oncologia Medica ,post-translational ,Immunology ,Population ,Synthetic lethality ,Article ,Cellular and Molecular Neuroscience ,Cancer stem cell ,chromatin ,colorectal neoplasms ,humans ,mitogen-activated protein kinase 14 ,neoplastic stem cells ,organoids ,prognosis ,protein processing, post-translational ,beta catenin ,Medicine ,Kinase activity ,colon cancer, p38, cancer stem cells ,lcsh:QH573-671 ,education ,Trametinib ,Settore MED/04 - Patologia Generale ,education.field_of_study ,business.industry ,lcsh:Cytology ,Cancer stem cells ,Wnt signaling pathway ,protein processing ,Cell Biology ,Colorectal cancer ,digestive system diseases ,Settore BIO/12 - Biochimica Clinica E Biologia Molecolare Clinica ,Catenin ,Cancer research ,Stem cell ,Settore MED/46 - Scienze Tecniche Di Medicina Di Laboratorio ,business ,Protein Processing, Post-Translational ,Post-translational modifications - Abstract
The prognosis of locally advanced colorectal cancer (CRC) is currently unsatisfactory. This is mainly due to drug resistance, recurrence, and subsequent metastatic dissemination, which are sustained by the cancer stem cell (CSC) population. The main driver of the CSC gene expression program is Wnt signaling, and previous reports indicate that Wnt3a can activate p38 MAPK. Besides, p38 was shown to feed into the canonical Wnt/β-catenin pathway. Here we show that patient-derived locally advanced CRC stem cells (CRC-SCs) are characterized by increased expression of p38α and are “addicted” to its kinase activity. Of note, we found that stage III CRC patients with high p38α levels display reduced disease-free and progression-free survival. Extensive molecular analysis in patient-derived CRC-SC tumorspheres and APCMin/+ mice intestinal organoids revealed that p38α acts as a β-catenin chromatin-associated kinase required for the regulation of a signaling platform involved in tumor proliferation, metastatic dissemination, and chemoresistance in these CRC model systems. In particular, the p38α kinase inhibitor ralimetinib, which has already entered clinical trials, promoted sensitization of patient-derived CRC-SCs to chemotherapeutic agents commonly used for CRC treatment and showed a synthetic lethality effect when used in combination with the MEK1 inhibitor trametinib. Taken together, these results suggest that p38α may be targeted in CSCs to devise new personalized CRC treatment strategies.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.