1
|
Healy FM, Turner AL, Marensi V, MacEwan DJ. Mediating kinase activity in Ras-mutant cancer: potential for an individualised approach? Front Pharmacol 2024; 15:1441938. [PMID: 39372214 PMCID: PMC11450236 DOI: 10.3389/fphar.2024.1441938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
It is widely acknowledged that there is a considerable number of oncogenic mutations within the Ras superfamily of small GTPases which are the driving force behind a multitude of cancers. Ras proteins mediate a plethora of kinase pathways, including the MAPK, PI3K, and Ral pathways. Since Ras was considered undruggable until recently, pharmacological targeting of pathways downstream of Ras has been attempted to varying success, though drug resistance has often proven an issue. Nuances between kinase pathway activation in the presence of various Ras mutants are thought to contribute to the resistance, however, the reasoning behind activation of different pathways in different Ras mutational contexts is yet to be fully elucidated. Indeed, such disparities often depend on cancer type and disease progression. However, we are in a revolutionary age of Ras mutant targeted therapy, with direct-targeting KRAS-G12C inhibitors revolutionising the field and achieving FDA-approval in recent years. However, these are only beneficial in a subset of patients. Approximately 90% of Ras-mutant cancers are not KRAS-G12C mutant, and therefore raises the question as to whether other distinct amino acid substitutions within Ras may one day be targetable in a similar manner, and indeed whether better understanding of the downstream pathways these various mutants activate could further improve therapy. Here, we discuss the favouring of kinase pathways across an array of Ras-mutant oncogenic contexts and assess recent advances in pharmacological targeting of various Ras mutants. Ultimately, we will examine the utility of individualised pharmacological approaches to Ras-mediated cancer.
Collapse
Affiliation(s)
- Fiona M. Healy
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Amy L. Turner
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Vanessa Marensi
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Chester Medical School, University of Chester, Chester, United Kingdom
| | - David J. MacEwan
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
2
|
Alsharoh H, Chiroi P, Isachesku E, Tanasa RA, Pop OL, Pirlog R, Berindan-Neagoe I. Personalizing Therapy Outcomes through Mitogen-Activated Protein Kinase Pathway Inhibition in Non-Small Cell Lung Cancer. Biomedicines 2024; 12:1489. [PMID: 39062063 PMCID: PMC11275062 DOI: 10.3390/biomedicines12071489] [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: 04/29/2024] [Revised: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Lung cancer (LC) is a highly invasive malignancy and the leading cause of cancer-related deaths, with non-small cell lung cancer (NSCLC) as its most prevalent histological subtype. Despite all breakthroughs achieved in drug development, the prognosis of NSCLC remains poor. The mitogen-activated protein kinase signaling cascade (MAPKC) is a complex network of interacting molecules that can drive oncogenesis, cancer progression, and drug resistance when dysregulated. Over the past decades, MAPKC components have been used to design MAPKC inhibitors (MAPKCIs), which have shown varying efficacy in treating NSCLC. Thus, recent studies support the potential clinical use of MAPKCIs, especially in combination with other therapeutic approaches. This article provides an overview of the MAPKC and its inhibitors in the clinical management of NSCLC. It addresses the gaps in the current literature on different combinations of selective inhibitors while suggesting two particular therapy approaches to be researched in NSCLC: parallel and aggregate targeting of the MAPKC. This work also provides suggestions that could serve as a potential guideline to aid future research in MAPKCIs to optimize clinical outcomes in NSCLC.
Collapse
Affiliation(s)
- Hasan Alsharoh
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | - Paul Chiroi
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | - Ekaterina Isachesku
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | | | - Ovidiu-Laurean Pop
- Department of Morphology Sciences, University of Oradea, 410087 Oradea, Romania;
| | - Radu Pirlog
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (H.A.); (P.C.); (E.I.); (I.B.-N.)
| |
Collapse
|
3
|
Townley R, Deniaud A, Stacy KS, Torres CSR, Cheraghi F, Wicker NB, de la Cova CC. The E3/E4 ubiquitin ligase UFD-2 suppresses normal and oncogenic signaling mediated by a Raf ortholog in Caenorhabditis elegans. Sci Signal 2023; 16:eabq4355. [PMID: 37643243 PMCID: PMC10656100 DOI: 10.1126/scisignal.abq4355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/10/2023] [Indexed: 08/31/2023]
Abstract
Signaling by the kinase cascade composed of Raf, MEK, and ERK is critical for animal development and is often inappropriately activated in human malignancies. We sought to identify factors that control signaling mediated by the Caenorhabditis elegans Raf ortholog LIN-45. A genetic screen showed that the degradation of LIN-45 required the E3/E4 ubiquitin ligase UFD-2. Both UFD-2 and its partner, the ATP-dependent segregase CDC-48, were required for the developmental regulation of LIN-45 protein abundance. We showed that UFD-2 acted in the same pathway as the E3 ubiquitin ligase SCFSEL-10 to decrease LIN-45 abundance in cells in which Raf-MEK-ERK signaling was most highly active. UFD-2 also reduced the protein abundance of activated LIN-45 carrying a mutation equivalent to the cancer-associated BRAF(V600E) variant. Our structure-function studies showed that the disruption of LIN-45 domains that mediate protein-protein interactions, including the conserved cysteine-rich domain and 14-3-3 binding motifs, were required for UFD-2-independent degradation of LIN-45. We propose a model in which UFD-2 and CDC-48 act downstream of SCFSEL-10 to remove LIN-45 from its protein interaction partners and facilitate proteasomal targeting and degradation. These findings imply that UFD-2 and CDC-48 may be important for Raf degradation during normal and oncogenic Ras and MAPK signaling in mammalian cells.
Collapse
Affiliation(s)
- Robert Townley
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, 53201 USA
| | - Augustin Deniaud
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, 53201 USA
| | - Kennedy S. Stacy
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, 53201 USA
| | | | - Fatemeh Cheraghi
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, 53201 USA
| | - Nicole B. Wicker
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, 53201 USA
| | - Claire C. de la Cova
- Department of Biological Sciences, University of Wisconsin-Milwaukee; Milwaukee, Wisconsin, 53201 USA
| |
Collapse
|
4
|
Sigaud R, Albert TK, Hess C, Hielscher T, Winkler N, Kocher D, Walter C, Münter D, Selt F, Usta D, Ecker J, Brentrup A, Hasselblatt M, Thomas C, Varghese J, Capper D, Thomale UW, Hernáiz Driever P, Simon M, Horn S, Herz NA, Koch A, Sahm F, Hamelmann S, Faria-Andrade A, Jabado N, Schuhmann MU, Schouten-van Meeteren AYN, Hoving E, Brummer T, van Tilburg CM, Pfister SM, Witt O, Jones DTW, Kerl K, Milde T. MAPK inhibitor sensitivity scores predict sensitivity driven by the immune infiltration in pediatric low-grade gliomas. Nat Commun 2023; 14:4533. [PMID: 37500667 PMCID: PMC10374577 DOI: 10.1038/s41467-023-40235-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
Pediatric low-grade gliomas (pLGG) show heterogeneous responses to MAPK inhibitors (MAPKi) in clinical trials. Thus, more complex stratification biomarkers are needed to identify patients likely to benefit from MAPKi therapy. Here, we identify MAPK-related genes enriched in MAPKi-sensitive cell lines using the GDSC dataset and apply them to calculate class-specific MAPKi sensitivity scores (MSSs) via single-sample gene set enrichment analysis. The MSSs discriminate MAPKi-sensitive and non-sensitive cells in the GDSC dataset and significantly correlate with response to MAPKi in an independent PDX dataset. The MSSs discern gliomas with varying MAPK alterations and are higher in pLGG compared to other pediatric CNS tumors. Heterogenous MSSs within pLGGs with the same MAPK alteration identify proportions of potentially sensitive patients. The MEKi MSS predicts treatment response in a small set of pLGG patients treated with trametinib. High MSSs correlate with a higher immune cell infiltration, with high expression in the microglia compartment in single-cell RNA sequencing data, while low MSSs correlate with low immune infiltration and increased neuronal score. The MSSs represent predictive tools for the stratification of pLGG patients and should be prospectively validated in clinical trials. Our data supports a role for microglia in the response to MAPKi.
Collapse
Affiliation(s)
- Romain Sigaud
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| | - Thomas K Albert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Caroline Hess
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Faculty of Biochemistry, Heidelberg University, Heidelberg, Germany
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Nadine Winkler
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Daniela Kocher
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Daniel Münter
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Florian Selt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Diren Usta
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jonas Ecker
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Angela Brentrup
- Neurosurgery Dept., University Hospital Münster, Münster, Germany
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Christian Thomas
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - David Capper
- Berlin Institute of Health, Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, Berlin, Germany
| | - Ulrich W Thomale
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Pediatric Neurosurgery, Berlin, Germany
| | - Pablo Hernáiz Driever
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, German HIT-LOGGIC-Registry for pLGG in children and adolescents, Department of Pediatric Oncology and Hematology, Berlin, Germany
| | - Michèle Simon
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, German HIT-LOGGIC-Registry for pLGG in children and adolescents, Department of Pediatric Oncology and Hematology, Berlin, Germany
| | - Svea Horn
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, German HIT-LOGGIC-Registry for pLGG in children and adolescents, Department of Pediatric Oncology and Hematology, Berlin, Germany
| | - Nina Annika Herz
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, German HIT-LOGGIC-Registry for pLGG in children and adolescents, Department of Pediatric Oncology and Hematology, Berlin, Germany
| | - Arend Koch
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, Berlin, Germany
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Hamelmann
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Nada Jabado
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, H4A 3J1, Canada
- Department of Pediatrics, McGill University, and The Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Martin U Schuhmann
- Section of Pediatric Neurosurgery, Department of Neurosurgery, University Hospital Tübingen, Tübingen, Germany
| | | | - Eelco Hoving
- Princess Màxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany, Centre for Biological Signaling Studies BIOSS, University of Freiburg and German Consortium for Translational Cancer Research (DKTK), Freiburg, Germany, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cornelis M van Tilburg
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
5
|
Oh KY, Kim JH, Cho SD, Yoon HJ, Lee JI, Hong SD. BRAF V600E and previously unidentified KRAS G12C mutations in odontogenic tumors may affect MAPK activation differently depending on tumor type. Genes Chromosomes Cancer 2022; 61:481-490. [PMID: 35353428 DOI: 10.1002/gcc.23040] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/10/2022] Open
Abstract
Although several types of odontogenic tumors share the same mutations in MAPK pathway genes, their effects on MAPK activation remain unclarified. This study aimed to evaluate the associations between these mutations and ERK phosphorylation in ameloblastoma and mixed odontogenic tumors (MOTs) and to analyze the expression pattern of phosphorylated ERK (p-ERK) for determining the involvement of MAPK activation in the development and progression of odontogenic tumors. Forty-three odontogenic tumors consisting of 18 ameloblastomas and 25 MOTs were analyzed for BRAF, KRAS, and NRAS mutations by Sanger sequencing. The expressions of BRAFV600E protein and p-ERK were detected by immunohistochemistry. The associations of mutation status and p-ERK expression were statistically analyzed. In ameloblastoma cells, the effect of BRAFV600E inhibition on MAPK activation was investigated. In benign MOTs, BRAFV600E mutations were neither expressed at the protein level nor associated with p-ERK expression. In contrast, BRAFV600E -mutant ameloblastic fibrosarcoma showed co-expression of BRAF V600E protein and p-ERK, especially in the sarcomatous component. In ameloblastoma, p-ERK was predominantly expressed in the tumor periphery showing a significant correlation with BRAFV600E mutations, and in vitro BRAFV600E inhibition decreased ERK phosphorylation. KRASG12C mutations, previously unidentified in odontogenic tumors, were detected in one case each of benign MOT and ameloblastoma; only the latter was high-p-ERK. In conclusion, unlike in benign MOTs, BRAFV600E and KRASG12C mutations lead to MAPK activation in ameloblastoma, suggesting their role as therapeutic targets. p-ERK intratumoral heterogeneity indicates that MAPK pathway activation may be associated with sarcomatous proliferation of ameloblastic fibrosarcoma and infiltrative behavior of ameloblastoma. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kyu-Young Oh
- Department of Oral Pathology, Seoul National University Dental Hospital, Seoul, Republic of Korea.,Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Ji-Hoon Kim
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, Seoul National University Dental Hospital, Seoul, Republic of Korea.,Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Hye-Jung Yoon
- Department of Oral Pathology, Seoul National University Dental Hospital, Seoul, Republic of Korea.,Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Jae-Il Lee
- Department of Oral Pathology, Seoul National University Dental Hospital, Seoul, Republic of Korea.,Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Seong-Doo Hong
- Department of Oral Pathology, Seoul National University Dental Hospital, Seoul, Republic of Korea.,Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Riobello C, Casanueva Muruais R, Suárez-Fernández L, García-Marín R, Cabal VN, Blanco-Lorenzo V, Franchi A, Laco J, López F, Llorente JL, Hermsen MA. Intragenic NF1 deletions in sinonasal mucosal malignant melanoma. Pigment Cell Melanoma Res 2021; 35:88-96. [PMID: 34547192 DOI: 10.1111/pcmr.13015] [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: 06/01/2021] [Revised: 08/27/2021] [Accepted: 09/18/2021] [Indexed: 11/29/2022]
Abstract
Mucosal malignant melanoma (MMM) is a rare and aggressive tumor. Despite effective local therapies, tumor recurrence and metastasis remain frequent. The genetics of MMM remain incompletely understood. This study is aimed to identify actionable genetic alterations by next-generation sequencing. Fifteen MMM samples were analyzed by next-generation and Sanger sequencing. Gene copy number alterations were analyzed by MLPA. Mutation status was correlated with pERK, pAKT, and Ki-67 expression and follow-up data. Inactivating mutations and intragenic deletions in neurofibromatosis type-1 (NF1) were identified in 3 and 2 cases, respectively, (in total 5/15, 33%) and activating mutations in NRAS and KRAS (3/15, 20%) cases. Other mutated genes included CDKN2A, APC, ATM, MITF, FGFR1, and FGFR2. BRAF and KIT mutations were not observed. Cases with NF1 alterations tended to have worse overall survival. The mutational status was not associated with pERK, pAKT, or Ki-67 immunostaining. MMM carries frequent gene mutations activating the MAPK pathway, similar to cutaneous melanoma. In contrast, NF1 is the most frequently affected gene. Intragenic NF1 deletions have not been described before and may go undetected by sequencing studies. This finding is clinically relevant as NF1-mutated melanomas have worse survival and could benefit from therapy with immune checkpoint and MEK inhibitors.
Collapse
Affiliation(s)
- Cristina Riobello
- Department Head and Neck Cancer, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Centro de Investigación Biomédica en Red (CIBER-ONC), Oviedo, Spain
| | | | - Laura Suárez-Fernández
- Department Head and Neck Cancer, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Centro de Investigación Biomédica en Red (CIBER-ONC), Oviedo, Spain
| | - Rocío García-Marín
- Department Head and Neck Cancer, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Centro de Investigación Biomédica en Red (CIBER-ONC), Oviedo, Spain
| | - Virginia N Cabal
- Department Head and Neck Cancer, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Centro de Investigación Biomédica en Red (CIBER-ONC), Oviedo, Spain
| | | | - Alessandro Franchi
- Department of Translational Research and of New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Jan Laco
- The Fingerland Dept Pathology, Charles University Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Fernando López
- Department Otolaryngology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - José Luis Llorente
- Department Otolaryngology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Mario A Hermsen
- Department Head and Neck Cancer, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Centro de Investigación Biomédica en Red (CIBER-ONC), Oviedo, Spain
| |
Collapse
|
7
|
Appleton KM, Palsuledesai CC, Misek SA, Blake M, Zagorski J, Gallo KA, Dexheimer TS, Neubig RR. Inhibition of the Myocardin-Related Transcription Factor Pathway Increases Efficacy of Trametinib in NRAS-Mutant Melanoma Cell Lines. Cancers (Basel) 2021; 13:cancers13092012. [PMID: 33921974 PMCID: PMC8122681 DOI: 10.3390/cancers13092012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Malignant melanoma is the most aggressive skin cancer, and treatment is often ineffective due to the development of resistance to targeted therapeutic agents. The most prevalent form of melanoma with a mutated BRAF gene has an effective treatment, but the second most common mutation in melanoma (NRAS) leads to tumors that lack targeted therapies. In this study, we show that NRAS mutant human melanoma cells that are most resistant to inhibition of the oncogenic pathway have a second activated pathway (Rho). Inhibiting that pathway at one of several points can produce more effective cell killing than inhibition of the NRAS pathway alone. This raises the possibility that such a combination treatment could prove effective in those melanomas that fail to respond to existing targeted therapies such as vemurafenib and trametinib. Abstract The Ras/MEK/ERK pathway has been the primary focus of targeted therapies in melanoma; it is aberrantly activated in almost 80% of human cutaneous melanomas (≈50% BRAFV600 mutations and ≈30% NRAS mutations). While drugs targeting the MAPK pathway have yielded success in BRAFV600 mutant melanoma patients, such therapies have been ineffective in patients with NRAS mutant melanomas in part due to their cytostatic effects and primary resistance. Here, we demonstrate that increased Rho/MRTF-pathway activation correlates with high intrinsic resistance to the MEK inhibitor, trametinib, in a panel of NRAS mutant melanoma cell lines. A combination of trametinib with the Rho/MRTF-pathway inhibitor, CCG-222740, synergistically reduced cell viability in NRAS mutant melanoma cell lines in vitro. Furthermore, the combination of CCG-222740 with trametinib induced apoptosis and reduced clonogenicity in SK-Mel-147 cells, which are highly resistant to trametinib. These findings suggest a role of the Rho/MRTF-pathway in intrinsic trametinib resistance in a subset of NRAS mutant melanoma cell lines and highlight the therapeutic potential of concurrently targeting the Rho/MRTF-pathway and MEK in NRAS mutant melanomas.
Collapse
Affiliation(s)
- Kathryn M. Appleton
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA; (K.M.A.); (C.C.P.); (M.B.); (J.Z.); (T.S.D.)
| | - Charuta C. Palsuledesai
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA; (K.M.A.); (C.C.P.); (M.B.); (J.Z.); (T.S.D.)
| | - Sean A. Misek
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (S.A.M.); (K.A.G.)
| | - Maja Blake
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA; (K.M.A.); (C.C.P.); (M.B.); (J.Z.); (T.S.D.)
| | - Joseph Zagorski
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA; (K.M.A.); (C.C.P.); (M.B.); (J.Z.); (T.S.D.)
| | - Kathleen A. Gallo
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (S.A.M.); (K.A.G.)
| | - Thomas S. Dexheimer
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA; (K.M.A.); (C.C.P.); (M.B.); (J.Z.); (T.S.D.)
| | - Richard R. Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA; (K.M.A.); (C.C.P.); (M.B.); (J.Z.); (T.S.D.)
- Department of Medicine, Division of Dermatology, Michigan State University, East Lansing, MI 48824, USA
- Correspondence: ; Tel.: +1-517-353-7145
| |
Collapse
|
8
|
Sun D, Li WY, Chen SH, Zhi ZF, Lin HS, Fan JT, Fan YJ. shRNA-Mediated Suppression of γ-Synuclein Leading to Downregulation of p38/ERK/JNK Phosphorylation and Cell Cycle Arrest in Endometrial Cancer Cells. Mol Biol 2021. [DOI: 10.1134/s0026893320060114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
9
|
Watanabe S, Shimomura A, Kubo T, Sekimizu M, Seo T, Watanabe SI, Kawai A, Yamamoto N, Tamura K, Kohno T, Ichikawa H, Yoshida A. BRAF V600E mutation is a potential therapeutic target for a small subset of synovial sarcoma. Mod Pathol 2020; 33:1660-1668. [PMID: 32238877 DOI: 10.1038/s41379-020-0530-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/12/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
Synovial sarcoma (SS) is an aggressive tumor that most often affects the deep soft tissues in young adults. Intrathoracic SS is rare and is associated with poor outcome, highlighting the urgent need for a novel therapeutic strategy. In the process of clinical sequencing, we identified two patients with intrathoracic SS harboring the BRAF V600E mutation. The patients were women aged 32 and 23 years, and both presented with SS18-SSX2-positive monophasic SS in the thoracic cavity. BRAF V600E mutations were detected by next generation sequencing, and validated immunohistochemically by diffuse intense positivity to BRAF V600E mutation-specific antibodies. The phosphorylated ERK (pERK) immunohistochemistry result was also positive. One patient received a combination therapy of dabrafenib and trametinib, which led to tumor shrinkage. However, the tumor growth progressed 7.5 months later with an additional NRAS Q61K mutation. Immunohistochemical screening of 67 archival SS tumor samples failed to identify additional samples with BRAF V600E mutation. However, 32% of BRAF V600E-negative cases was positive for pERK, and one of the six tumors showing the highest pERK expression harbored an FGFR2-activating mutation. This is the first report of targetable BRAF mutation in a small subset of SS. Our study suggests involvement of the mitogen-activated protein kinase pathway and the potential clinical implication of BRAF mutation screening in SS.
Collapse
Affiliation(s)
- Sho Watanabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan.,Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center, National Cancer Center East, Chiba, Japan
| | - Akihiko Shimomura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Kubo
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan.,Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Masaya Sekimizu
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan.,Department of Orthopaedic Surgery, Showa University School of Medicine, Tokyo, Japan
| | - Takuji Seo
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shun-Ichi Watanabe
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan.,Rare Cancer Center, National Cancer Center Hospital, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Kohno
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan.,Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hitoshi Ichikawa
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan.,Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Akihiko Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan. .,Rare Cancer Center, National Cancer Center Hospital, Tokyo, Japan.
| |
Collapse
|
10
|
Cronise KE, Hernandez BG, Gustafson DL, Duval DL. Identifying the ErbB/MAPK Signaling Cascade as a Therapeutic Target in Canine Bladder Cancer. Mol Pharmacol 2019; 96:36-46. [PMID: 31048548 DOI: 10.1124/mol.119.115808] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/27/2019] [Indexed: 12/19/2022] Open
Abstract
Transitional cell carcinoma (TCC) of the bladder comprises 2% of diagnosed canine cancers. TCC tumors are generally inoperable and unresponsive to traditional chemotherapy, indicating a need for more effective therapies. BRAF, a kinase in the mitogen-activated protein kinase (MAPK) pathway, is mutated in 70% of canine TCCs. In this study, we use BRAF mutant and wild-type TCC cell lines to characterize the role of BRAF mutations in TCC pathogenesis and assess the efficacy of inhibition of the MAPK pathway alone and in combination with other gene targets as a treatment for canine TCC. Analysis of MAPK target gene expression and assessment of extracellular signal-regulated kinase (ERK) 1/2 phosphorylation following serum starvation indicated constitutive MAPK activity in all TCC cell lines. BRAF mutant TCC cell lines were insensitive to the BRAF inhibitor vemurafenib, with IC50 values greater than 5 μM, but exhibited greater sensitivity to a paradox-breaking BRAF inhibitor (IC50: 0.2-1 μM). All TCC cell lines had IC50 values less than 7 nM to the mitogen-activated protein kinase kinase (MEK) 1/2 inhibitor trametinib independent of their BRAF mutation status. ERK1/2 phosphorylation decreased after 6-hour treatments with MAPK inhibitors, but rebounded by 24 hours, suggesting the presence of resistance mechanisms. Microarray analysis identified elevated expression of the ErbB family of receptors and ligands in TCC cell lines. The pan-ErbB inhibitor sapitinib synergized with BRAF inhibition in BRAF mutant Bliley TCC cells and synergized with MEK1/2 inhibition in Bliley and BRAF wild-type Kinsey cells. These findings suggest the potential for combined MAPK and ErbB receptor inhibition as a therapy for canine TCC. SIGNIFICANCE STATEMENT: The results of this study (1) identify a novel combination strategy for canine bladder cancer treatment: targeting the ErbB/MAPK signaling cascade and (2) establish the utility of canine bladder cancer as a naturally-occurring model for human MAPK-driven cancers.
Collapse
Affiliation(s)
- Kathryn E Cronise
- Flint Animal Cancer Center, Department of Clinical Sciences (K.E.C., B.G.H., D.L.G., D.L.D.), and Cell and Molecular Biology Graduate Program (K.E.C., D.L.G., D.L.D.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G., D.L.D.)
| | - Belen G Hernandez
- Flint Animal Cancer Center, Department of Clinical Sciences (K.E.C., B.G.H., D.L.G., D.L.D.), and Cell and Molecular Biology Graduate Program (K.E.C., D.L.G., D.L.D.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G., D.L.D.)
| | - Daniel L Gustafson
- Flint Animal Cancer Center, Department of Clinical Sciences (K.E.C., B.G.H., D.L.G., D.L.D.), and Cell and Molecular Biology Graduate Program (K.E.C., D.L.G., D.L.D.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G., D.L.D.)
| | - Dawn L Duval
- Flint Animal Cancer Center, Department of Clinical Sciences (K.E.C., B.G.H., D.L.G., D.L.D.), and Cell and Molecular Biology Graduate Program (K.E.C., D.L.G., D.L.D.), Colorado State University, Fort Collins, Colorado; and University of Colorado Cancer Center, Aurora, Colorado (D.L.G., D.L.D.)
| |
Collapse
|
11
|
Shain AH, Joseph NM, Yu R, Benhamida J, Liu S, Prow T, Ruben B, North J, Pincus L, Yeh I, Judson R, Bastian BC. Genomic and Transcriptomic Analysis Reveals Incremental Disruption of Key Signaling Pathways during Melanoma Evolution. Cancer Cell 2018; 34:45-55.e4. [PMID: 29990500 PMCID: PMC6319271 DOI: 10.1016/j.ccell.2018.06.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/02/2018] [Accepted: 06/08/2018] [Indexed: 12/30/2022]
Abstract
We elucidated genomic and transcriptomic changes that accompany the evolution of melanoma from pre-malignant lesions by sequencing DNA and RNA from primary melanomas and their adjacent precursors, as well as matched primary tumors and regional metastases. In total, we analyzed 230 histopathologically distinct areas of melanocytic neoplasia from 82 patients. Somatic alterations sequentially induced mitogen-activated protein kinase (MAPK) pathway activation, upregulation of telomerase, modulation of the chromatin landscape, G1/S checkpoint override, ramp-up of MAPK signaling, disruption of the p53 pathway, and activation of the PI3K pathway; no mutations were specifically associated with metastatic progression, as these pathways were perturbed during the evolution of primary melanomas. UV radiation-induced point mutations steadily increased until melanoma invasion, at which point copy-number alterations also became prevalent.
Collapse
Affiliation(s)
- A Hunter Shain
- University of California San Francisco, Department of Dermatology, San Francisco, CA, USA; University of California San Francisco, Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA.
| | - Nancy M Joseph
- University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| | - Richard Yu
- University of California San Francisco, Department of Dermatology, San Francisco, CA, USA; University of California San Francisco, Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Jamal Benhamida
- University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| | - Shanshan Liu
- University of California San Francisco, Department of Dermatology, San Francisco, CA, USA; University of California San Francisco, Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Tarl Prow
- Future Industries Institute, University of South Australia, Adelaide, SA, Australia
| | - Beth Ruben
- University of California San Francisco, Department of Dermatology, San Francisco, CA, USA; University of California San Francisco, Department of Pathology, San Francisco, CA, USA; Palo Alto Medical Foundation, Palo Alto, CA, USA
| | - Jeffrey North
- University of California San Francisco, Department of Dermatology, San Francisco, CA, USA; University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| | - Laura Pincus
- University of California San Francisco, Department of Dermatology, San Francisco, CA, USA; University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| | - Iwei Yeh
- University of California San Francisco, Department of Dermatology, San Francisco, CA, USA; University of California San Francisco, Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA; University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| | - Robert Judson
- University of California San Francisco, Department of Dermatology, San Francisco, CA, USA; University of California San Francisco, Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Boris C Bastian
- University of California San Francisco, Department of Dermatology, San Francisco, CA, USA; University of California San Francisco, Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA; University of California San Francisco, Department of Pathology, San Francisco, CA, USA.
| |
Collapse
|
12
|
Wagle MC, Kirouac D, Klijn C, Liu B, Mahajan S, Junttila M, Moffat J, Merchant M, Huw L, Wongchenko M, Okrah K, Srinivasan S, Mounir Z, Sumiyoshi T, Haverty PM, Yauch RL, Yan Y, Kabbarah O, Hampton G, Amler L, Ramanujan S, Lackner MR, Huang SMA. A transcriptional MAPK Pathway Activity Score (MPAS) is a clinically relevant biomarker in multiple cancer types. NPJ Precis Oncol 2018; 2:7. [PMID: 29872725 PMCID: PMC5871852 DOI: 10.1038/s41698-018-0051-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/29/2018] [Accepted: 02/05/2018] [Indexed: 12/18/2022] Open
Abstract
KRAS- and BRAF-mutant tumors are often dependent on MAPK signaling for proliferation and survival and thus sensitive to MAPK pathway inhibitors. However, clinical studies have shown that MEK inhibitors are not uniformly effective in these cancers indicating that mutational status of these oncogenes does not accurately capture MAPK pathway activity. A number of transcripts are regulated by this pathway and are recurrently identified in genome-based MAPK transcriptional signatures. To test whether the transcriptional output of only 10 of these targets could quantify MAPK pathway activity with potential predictive or prognostic clinical utility, we created a MAPK Pathway Activity Score (MPAS) derived from aggregated gene expression. In vitro, MPAS predicted sensitivity to MAPK inhibitors in multiple cell lines, comparable to or better than larger genome-based statistical models. Bridging in vitro studies and clinical samples, median MPAS from a given tumor type correlated with cobimetinib (MEK inhibitor) sensitivity of cancer cell lines originating from the same tissue type. Retrospective analyses of clinical datasets showed that MPAS was associated with the sensitivity of melanomas to vemurafenib (HR: 0.596) and negatively prognostic of overall or progression-free survival in both adjuvant and metastatic CRC (HR: 1.5 and 1.4), adrenal cancer (HR: 1.7), and HER2+ breast cancer (HR: 1.6). MPAS thus demonstrates potential clinical utility that warrants further exploration.
Collapse
Affiliation(s)
- Marie-Claire Wagle
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Daniel Kirouac
- 2Department of Pre-Clinical and Translational PKPD, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Christiaan Klijn
- 3Department of Bioinformatics, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Bonnie Liu
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Shilpi Mahajan
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Melissa Junttila
- 4Department of Translational Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - John Moffat
- 5Department of Biochemical and Cellular pharmacology, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Mark Merchant
- 4Department of Translational Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Ling Huw
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Matthew Wongchenko
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Kwame Okrah
- 6Department of Biostatistics, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Shrividhya Srinivasan
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Zineb Mounir
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Teiko Sumiyoshi
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Peter M Haverty
- 3Department of Bioinformatics, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Robert L Yauch
- 4Department of Translational Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Yibing Yan
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Omar Kabbarah
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Garret Hampton
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Lukas Amler
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Saroja Ramanujan
- 2Department of Pre-Clinical and Translational PKPD, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Mark R Lackner
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Shih-Min A Huang
- 1Department of Oncology Biomarker Development, Genentech, 1 DNA Way, South San Francisco, CA 94080 USA.,7Present Address: Bristol-Myers Squibb, 3551 Lawrenceville Princeton, Lawrence Township, NJ 08648 USA
| |
Collapse
|
13
|
Moschos SJ, Sullivan RJ, Hwu WJ, Ramanathan RK, Adjei AA, Fong PC, Shapira-Frommer R, Tawbi HA, Rubino J, Rush TS, Zhang D, Miselis NR, Samatar AA, Chun P, Rubin EH, Schiller J, Long BJ, Dayananth P, Carr D, Kirschmeier P, Bishop WR, Deng Y, Cooper A, Shipps GW, Moreno BH, Robert L, Ribas A, Flaherty KT. Development of MK-8353, an orally administered ERK1/2 inhibitor, in patients with advanced solid tumors. JCI Insight 2018; 3:92352. [PMID: 29467321 DOI: 10.1172/jci.insight.92352] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 12/28/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Constitutive activation of ERK1/2 occurs in various cancers, and its reactivation is a well-described resistance mechanism to MAPK inhibitors. ERK inhibitors may overcome the limitations of MAPK inhibitor blockade. The dual mechanism inhibitor SCH772984 has shown promising preclinical activity across various BRAFV600/RAS-mutant cancer cell lines and human cancer xenografts. METHODS We have developed an orally bioavailable ERK inhibitor, MK-8353; conducted preclinical studies to demonstrate activity, pharmacodynamic endpoints, dosing, and schedule; completed a study in healthy volunteers (P07652); and subsequently performed a phase I clinical trial in patients with advanced solid tumors (MK-8353-001). In the P07652 study, MK-8353 was administered as a single dose in 10- to 400-mg dose cohorts, whereas in the MK-8353-001 study, MK-8353 was administered in 100- to 800-mg dose cohorts orally twice daily. Safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity were analyzed. RESULTS MK-8353 exhibited comparable potency with SCH772984 across various preclinical cancer models. Forty-eight patients were enrolled in the P07652 study, and twenty-six patients were enrolled in the MK-8353-001 study. Adverse events included diarrhea (44%), fatigue (40%), nausea (32%), and rash (28%). Dose-limiting toxicity was observed in the 400-mg and 800-mg dose cohorts. Sufficient exposure to MK-8353 was noted that correlated with biological activity in preclinical data. Three of fifteen patients evaluable for treatment response in the MK-8353-001 study had partial response, all with BRAFV600-mutant melanomas. CONCLUSION MK-8353 was well tolerated up to 400 mg twice daily and exhibited antitumor activity in patients with BRAFV600-mutant melanoma. However, antitumor activity was not particularly correlated with pharmacodynamic parameters. TRIAL REGISTRATION ClinicalTrials.gov NCT01358331. FUNDING Merck Sharp & Dohme Corp., a subsidiary of Merck & Co. Inc., and NIH (P01 CA168585 and R35 CA197633).
Collapse
Affiliation(s)
- Stergios J Moschos
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ramesh K Ramanathan
- Translational Genomics Research Institute, Phoenix, Arizona, USA; Virginia G. Piper Cancer Center, Scottsdale, Arizona, USA
| | - Alex A Adjei
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Peter C Fong
- The University of Auckland and Auckland City Hospital, Auckland, New Zealand
| | | | - Hussein A Tawbi
- University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | | | | | - Da Zhang
- Merck & Co. Inc., Kenilworth, New Jersey, USA
| | | | | | | | | | | | | | | | - Donna Carr
- Merck & Co. Inc., Kenilworth, New Jersey, USA
| | | | | | - Yongqi Deng
- Merck & Co. Inc., Kenilworth, New Jersey, USA
| | - Alan Cooper
- Merck & Co. Inc., Kenilworth, New Jersey, USA
| | | | - Blanca Homet Moreno
- Jonsson Comprehensive Cancer Center at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Lidia Robert
- Jonsson Comprehensive Cancer Center at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center at UCLA, University of California Los Angeles, Los Angeles, California, USA
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Kiniwa Y, Nakamura K, Mikoshiba A, Akiyama Y, Morimoto A, Okuyama R. Diversity of circulating tumor cells in peripheral blood: Detection of heterogeneous BRAF mutations in a patient with advanced melanoma by single-cell analysis. J Dermatol Sci 2018; 90:211-213. [PMID: 29426605 DOI: 10.1016/j.jdermsci.2018.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/05/2018] [Accepted: 01/18/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Yukiko Kiniwa
- Department of Dermatology, Shinshu University School of Medicine, Japan
| | - Kenta Nakamura
- Department of Dermatology, Shinshu University School of Medicine, Japan
| | - Asuka Mikoshiba
- Department of Dermatology, Shinshu University School of Medicine, Japan
| | | | | | - Ryuhei Okuyama
- Department of Dermatology, Shinshu University School of Medicine, Japan.
| |
Collapse
|
15
|
Rodríguez CI, Castro-Pérez E, Prabhakar K, Block L, Longley BJ, Wisinski JA, Kimple ME, Setaluri V. EPAC-RAP1 Axis-Mediated Switch in the Response of Primary and Metastatic Melanoma to Cyclic AMP. Mol Cancer Res 2017; 15:1792-1802. [PMID: 28851815 PMCID: PMC6309370 DOI: 10.1158/1541-7786.mcr-17-0067] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/30/2017] [Accepted: 08/23/2017] [Indexed: 11/16/2022]
Abstract
Cyclic AMP (cAMP) is an important second messenger that regulates a wide range of physiologic processes. In mammalian cutaneous melanocytes, cAMP-mediated signaling pathways activated by G-protein-coupled receptors (GPCR), like melanocortin 1 receptor (MC1R), play critical roles in melanocyte homeostasis including cell survival, proliferation, and pigment synthesis. Impaired cAMP signaling is associated with increased risk of cutaneous melanoma. Although mutations in MAPK pathway components are the most frequent oncogenic drivers of melanoma, the role of cAMP in melanoma is not well understood. Here, using the Braf(V600E)/Pten-null mouse model of melanoma, topical application of an adenylate cyclase agonist, forskolin (a cAMP inducer), accelerated melanoma tumor development in vivo and stimulated the proliferation of mouse and human primary melanoma cells, but not human metastatic melanoma cells in vitro The differential response of primary and metastatic melanoma cells was also evident upon pharmacologic inhibition of the cAMP effector protein kinase A. Pharmacologic inhibition and siRNA-mediated knockdown of other cAMP signaling pathway components showed that EPAC-RAP1 axis, an alternative cAMP signaling pathway, mediates the switch in response of primary and metastatic melanoma cells to cAMP. Evaluation of pERK levels revealed that this phenotypic switch was not correlated with changes in MAPK pathway activity. Although cAMP elevation did not alter the sensitivity of metastatic melanoma cells to BRAF(V600E) and MEK inhibitors, the EPAC-RAP1 axis appears to contribute to resistance to MAPK pathway inhibition. These data reveal a MAPK pathway-independent switch in response to cAMP signaling during melanoma progression.Implications: The prosurvival mechanism involving the cAMP-EPAC-RAP1 signaling pathway suggest the potential for new targeted therapies in melanoma. Mol Cancer Res; 15(12); 1792-802. ©2017 AACR.
Collapse
Affiliation(s)
- Carlos I Rodríguez
- Molecular and Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Edgardo Castro-Pérez
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Kirthana Prabhakar
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Laura Block
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - B Jack Longley
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Jaclyn A Wisinski
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Michelle E Kimple
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin
- Department of Medicine, Division of Endocrinology, School of Medicine and Public Health, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Vijayasaradhi Setaluri
- Molecular and Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.
- Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| |
Collapse
|
16
|
The impact of melanoma genetics on treatment response and resistance in clinical and experimental studies. Cancer Metastasis Rev 2017; 36:53-75. [PMID: 28210865 DOI: 10.1007/s10555-017-9657-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Recent attempts to characterize the melanoma mutational landscape using high-throughput sequencing technologies have identified new genes and pathways involved in the molecular pathogenesis of melanoma. Apart from mutated BRAF, NRAS, and KIT, a series of new recurrently mutated candidate genes with impact on signaling pathways have been identified such as NF1, PTEN, IDH1, RAC1, ARID2, and TP53. Under targeted treatment using BRAF and MEK1/2 inhibitors either alone or in combination, a majority of patients experience recurrences, which are due to different genetic mechanisms such as gene amplifications of BRAF or NRAS, MEK1/2 and PI3K mutations. In principle, resistance mechanisms converge on two signaling pathways, MAPK and PI3K-AKT-mTOR pathways. Resistance may be due to small subsets of resistant cells within a heterogeneous tumor mass not identified by sequencing of the bulk tumor. Future sequencing studies addressing tumor heterogeneity, e.g., by using single-cell sequencing technology, will most likely improve this situation. Gene expression patterns of metastatic lesions were also shown to predict treatment response, e.g., a MITF-low/NF-κB-high melanoma phenotype is resistant against classical targeted therapies. Finally, more recent treatment approaches using checkpoint inhibitors directed against PD-1 and CTLA-4 are very effective in melanoma and other tumor entities. Here, the mutational and neoantigen load of melanoma lesions may help to predict treatment response. Taken together, the new sequencing, molecular, and bioinformatic technologies exploiting the melanoma genome for treatment decisions have significantly improved our understanding of melanoma pathogenesis, treatment response, and resistance for either targeted treatment or immune checkpoint blockade.
Collapse
|
17
|
Shi SS, Wang X, Xia QY, Rao Q, Shen Q, Ye SB, Li R, Shi QL, Lu ZF, Ma HH, Zhou XJ. P16 overexpression inBRAF-mutated gastrointestinal stromal tumors. Expert Rev Mol Diagn 2016; 17:195-201. [PMID: 28034324 DOI: 10.1080/14737159.2017.1272413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Shan-shan Shi
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| | - Xuan Wang
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| | - Qiu-yuan Xia
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| | - Qiu Rao
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| | - Qin Shen
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| | - Sheng-bin Ye
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| | - Rui Li
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| | - Qun-li Shi
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| | - Zhen-feng Lu
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| | - Heng-hui Ma
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| | - Xiao-jun Zhou
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, P. R. China
| |
Collapse
|
18
|
Kawabata N, Matsuda M. Cell Density-Dependent Increase in Tyrosine-Monophosphorylated ERK2 in MDCK Cells Expressing Active Ras or Raf. PLoS One 2016; 11:e0167940. [PMID: 27936234 PMCID: PMC5148048 DOI: 10.1371/journal.pone.0167940] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/22/2016] [Indexed: 12/25/2022] Open
Abstract
The extracellular signal-regulated kinase (ERK) is one of the principal hub proteins that transmit growth signals from upstream oncogene products including Ras and BRaf to downstream effector proteins. However, there are both reports supporting and refuting the increase in ERK activity in cancer tissues expressing the active Ras and BRaf proteins. We considered that the cell density might account for this discrepancy. To examine this possibility, we prepared Madin-Darby canine kidney (MDCK) cells that expressed an active HRas, NRas, KRas, or BRaf and an ERK biosensor based on the principle of Förster resonance energy transfer (FRET). As we anticipated, expression of the active Ras or BRaf increased ERK activity at low cell densities. However, the ERK activity was markedly suppressed at high cell densities irrespective of the expression of the active Ras or BRaf. Western blotting analysis with Phos-tag gel revealed the decrease of tyrosine and threonine-diphosphorylated active ERK and the increase of tyrosine-monophosphorylated inactive ERK at high cell density. In addition, we found that calyculin A, an inhibitor for PPP-subfamily protein serine/threonine phosphatases, decreased the tyrosine-monophosphorylated ERK. Our study suggests that PPP-subfamily phosphatases may be responsible for cell density-dependent ERK dephosphorylation in cancer cells expressing active Ras or BRaf protein.
Collapse
Affiliation(s)
- Noriyuki Kawabata
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
19
|
Cao J, Heijkants RC, Jochemsen AG, Dogrusöz M, de Lange MJ, van der Velden PA, van der Burg SH, Jager MJ, Verdijk RM. Targeting of the MAPK and AKT pathways in conjunctival melanoma shows potential synergy. Oncotarget 2016; 8:58021-58036. [PMID: 28938534 PMCID: PMC5601630 DOI: 10.18632/oncotarget.10770] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 06/09/2016] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Conjunctival melanoma (CM) is a rare but lethal form of cancer. Similar to cutaneous melanoma, CM frequently carries activating mutations in BRAF and NRAS. We studied whether CM as well as conjunctival benign and premalignant melanocytic lesions express targets in the mitogen-activated protein kinase (MAPK) and AKT pathways, and whether specific inhibitors can suppress CM growth in vitro. METHODS 131 conjunctival lesions obtained from 129 patients were collected. The presence of BRAF V600E mutation and expression of phosphorylated (p)-ERK and p-AKT were assessed by immunohistochemistry. We studied cell proliferation, phosphorylation, cell cycling and apoptosis in three CM cell lines using two BRAF inhibitors (Vemurafenib and Dabrafenib), a MEK inhibitor (MEK162) and an AKT inhibitor (MK2206). RESULTS The BRAF V600E mutation was present in 19% of nevi and 26% of melanomas, but not in primary acquired melanosis (PAM). Nuclear and cytoplasmic p-ERK and p-AKT were expressed in all conjunctival lesions. Both BRAF inhibitors suppressed growth of both BRAF mutant CM cell lines, but only one induced cell death. MEK162 and MK2206 inhibited proliferation of CM cells in a dose-dependent manner, and the combination of these two drugs led to synergistic growth inhibition and cell death in all CM cell lines. CONCLUSION ERK and AKT are constitutively activated in conjunctival nevi, PAM and melanoma. While BRAF inhibitors prohibited cell growth, they were not always cytotoxic. Combining MEK and AKT inhibitors led to more growth inhibition and cell death in CM cells. The combination may benefit patients suffering from metastatic conjunctival melanoma.
Collapse
Affiliation(s)
- Jinfeng Cao
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Renier C Heijkants
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Aart G Jochemsen
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mehmet Dogrusöz
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mark J de Lange
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert M Verdijk
- Department of Pathology, Section Ophthalmic Pathology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
20
|
Holck S, Bonde J, Pedersen H, Petersen AA, Chaube A, Nielsen HJ, Larsson LI. Localization of active, dually phosphorylated extracellular signal-regulated kinase 1 and 2 in colorectal cancer with or without activating BRAF and KRAS mutations. Hum Pathol 2016; 54:37-46. [PMID: 27036313 DOI: 10.1016/j.humpath.2016.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 02/24/2016] [Accepted: 03/01/2016] [Indexed: 01/31/2023]
Abstract
Colorectal cancers (CRC) often show activating mutations of the KRAS or BRAF genes, which stimulate the extracellular signal-regulated kinase (ERK) pathway, thus increasing cell proliferation and inhibiting apoptosis. However, immunohistochemical results on ERK activation in such tumors differ greatly. Recently, using a highly optimized immunohistochemical method, we obtained evidence that high levels of ERK activation in rectal adenocarcinomas were associated with resistance to radiochemotherapy. In order to determine whether KRAS and/or BRAF mutations correlate to immunohistochemically detectable increases in phosphorylation of ERK (pERK), we stained biopsies from 36 CRC patients with activating mutations in the BRAF gene (BRAFV600E: BRAF(m)), the KRAS gene (KRAS(m)) or in neither (BRAF/KRAS(n)) with this optimized method. Staining was scored in blind-coded specimens by two observers. Staining of stromal cells was used as a positive control. BRAF(m) or KRAS(m) tumors did not show higher staining scores than BRAF/KRAS(n) tumors. Although BRAFV600E staining occurred in over 90% of cancer cells in all 9 BRAF(m) tumors, 3 only showed staining for pERK in less than 10% of cancer cell nuclei. The same applied to 4 of the 14 KRAS(m) tumors. A phophorylation-insensitive antibody demonstrated that lack of pERK staining did not reflect defect expression of ERK1/2 protein. Thus, increased staining for pERK does not correlate to BRAF or KRAS mutations even with a highly optimized procedure. Further studies are required to determine whether this reflects differences in expression of counterregulatory molecules, including ERK phosphatases.
Collapse
Affiliation(s)
- Susanne Holck
- Department of Pathology, Copenhagen University Hospital, DK -2650, Hvidovre, Denmark
| | - Jesper Bonde
- Department of Pathology, Copenhagen University Hospital, DK -2650, Hvidovre, Denmark; Clinical Research Centre, Copenhagen University Hospital, DK -2650, Hvidovre, Denmark
| | - Helle Pedersen
- Department of Pathology, Copenhagen University Hospital, DK -2650, Hvidovre, Denmark
| | - Anja Alex Petersen
- Department of Pathology, Copenhagen University Hospital, DK -2650, Hvidovre, Denmark; Clinical Research Centre, Copenhagen University Hospital, DK -2650, Hvidovre, Denmark
| | - Amita Chaube
- Department of Pathology, Copenhagen University Hospital, DK -2650, Hvidovre, Denmark
| | - Hans Jørgen Nielsen
- Department of Surgical Gastroenterology, Copenhagen University Hospital, DK -2650, Hvidovre, Denmark
| | - Lars-Inge Larsson
- Department of Pathology, Copenhagen University Hospital, DK -2650, Hvidovre, Denmark; Clinical Research Centre, Copenhagen University Hospital, DK -2650, Hvidovre, Denmark.
| |
Collapse
|
21
|
Wellbrock C, Arozarena I. Microphthalmia-associated transcription factor in melanoma development and MAP-kinase pathway targeted therapy. Pigment Cell Melanoma Res 2015; 28:390-406. [PMID: 25818589 PMCID: PMC4692100 DOI: 10.1111/pcmr.12370] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/16/2015] [Indexed: 12/12/2022]
Abstract
Malignant melanoma is a neoplasm of melanocytes, and the microphthalmia-associated transcription factor (MITF) is essential for the existence of melanocytes. MITF's relevance for this cell lineage is maintained in melanoma, where it is an important regulator of survival and balances melanoma cell proliferation with terminal differentiation (pigmentation). The MITF gene is amplified in ~20% of melanomas and MITF mutation can predispose to melanoma development. Furthermore, the regulation of MITF expression and function is strongly linked to the BRAF/MEK/ERK/MAP-kinase (MAPK) pathway, which is deregulated in >90% of melanomas and central target of current therapies. MITF expression in melanoma is heterogeneous, and recent findings highlight the relevance of this heterogeneity for the response of melanoma to MAPK pathway targeting drugs, as well as for MITF's role in melanoma progression. This review aims to provide an updated overview on the regulation of MITF function and plasticity in melanoma with a focus on its link to MAPK signaling.
Collapse
Affiliation(s)
- Claudia Wellbrock
- Manchester Cancer Research CentreWellcome Trust Centre for Cell Matrix ResearchFaculty of Life SciencesThe University of ManchesterManchesterUK
| | - Imanol Arozarena
- Manchester Cancer Research CentreWellcome Trust Centre for Cell Matrix ResearchFaculty of Life SciencesThe University of ManchesterManchesterUK
| |
Collapse
|
22
|
Macerola E, Loggini B, Giannini R, Garavello G, Giordano M, Proietti A, Niccoli C, Basolo F, Fontanini G. Coexistence of TERT promoter and BRAF mutations in cutaneous melanoma is associated with more clinicopathological features of aggressiveness. Virchows Arch 2015; 467:177-84. [DOI: 10.1007/s00428-015-1784-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/09/2015] [Accepted: 05/04/2015] [Indexed: 10/23/2022]
|
23
|
Oyama S, Funasaka Y, Watanabe A, Takizawa T, Kawana S, Saeki H. BRAF,KITandNRASmutations and expression of c-KIT, phosphorylated extracellular signal-regulated kinase and phosphorylated AKT in Japanese melanoma patients. J Dermatol 2015; 42:477-84. [DOI: 10.1111/1346-8138.12822] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 01/03/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Satomi Oyama
- Department of Dermatology; Graduate School of Medicine; Nippon Medical School; Tokyo Japan
| | - Yoko Funasaka
- Department of Dermatology; Graduate School of Medicine; Nippon Medical School; Tokyo Japan
| | - Atsushi Watanabe
- Department of Molecular and Medical Genetics; Graduate School of Medicine; Nippon Medical School; Tokyo Japan
| | - Toshihiro Takizawa
- Department of Molecular Medicine and Anatomy; Graduate School of Medicine; Nippon Medical School; Tokyo Japan
| | - Seiji Kawana
- Department of Dermatology; Graduate School of Medicine; Nippon Medical School; Tokyo Japan
| | - Hidehisa Saeki
- Department of Dermatology; Graduate School of Medicine; Nippon Medical School; Tokyo Japan
| |
Collapse
|
24
|
Garay T, Molnár E, Juhász É, László V, Barbai T, Dobos J, Schelch K, Pirker C, Grusch M, Berger W, Tímár J, Hegedűs B. Sensitivity of Melanoma Cells to EGFR and FGFR Activation but Not Inhibition is Influenced by Oncogenic BRAF and NRAS Mutations. Pathol Oncol Res 2015; 21:957-68. [PMID: 25749811 DOI: 10.1007/s12253-015-9916-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/17/2015] [Indexed: 11/28/2022]
Abstract
BRAF and NRAS are the two most frequent oncogenic driver mutations in melanoma and are pivotal components of both the EGF and FGF signaling network. Accordingly, we investigated the effect of BRAF and NRAS oncogenic mutation on the response to the stimulation and inhibition of epidermal and fibroblast growth factor receptors in melanoma cells. In the three BRAF mutant, two NRAS mutant and two double wild-type cell lines growth factor receptor expression had been verified by qRT-PCR. Cell proliferation and migration were determined by the analysis of 3-days-long time-lapse videomicroscopic recordings. Of note, a more profound response was found in motility as compared to proliferation and double wild-type cells displayed a higher sensitivity to EGF and FGF2 treatment when compared to mutant cells. Both baseline and induced activation of the growth factor signaling was assessed by immunoblot analysis of the phosphorylation of the downstream effectors Erk1/2. Low baseline and higher inducibility of the signaling pathway was characteristic in double wild-type cells. In contrast, oncogenic BRAF or NRAS mutation did not influence the response to EGF or FGF receptor inhibitors in vitro. Our findings demonstrate that the oncogenic mutations in melanoma have a profound impact on the motogenic effect of the activation of growth factor receptor signaling. Since emerging molecularly targeted therapies aim at the growth factor receptor signaling, the appropriate mutational analysis of individual melanoma cases is essential in both preclinical studies and in the clinical trials and practice.
Collapse
Affiliation(s)
- Tamás Garay
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, H-1091, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
|
26
|
Lade-Keller J, Riber-Hansen R, Guldberg P, Schmidt H, Hamilton-Dutoit SJ, Steiniche T. Immunohistochemical analysis of molecular drivers in melanoma identifies p16 as an independent prognostic biomarker. J Clin Pathol 2014; 67:520-8. [DOI: 10.1136/jclinpath-2013-202127] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
27
|
Kunz M. Oncogenes in melanoma: an update. Eur J Cell Biol 2013; 93:1-10. [PMID: 24468268 DOI: 10.1016/j.ejcb.2013.12.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/16/2013] [Accepted: 12/17/2013] [Indexed: 12/13/2022] Open
Abstract
Melanoma is a highly aggressive tumour with poor prognosis in the metastatic stage. BRAF, NRAS, and KIT are three well-known oncogenes involved in melanoma pathogenesis. Targeting of mutated BRAF kinase has recently been shown to significantly improve overall survival of metastatic melanoma patients, underscoring the particular role of this oncogene in melanoma biology. However, recurrences regularly occur within several months, which supposedly involve further oncogenes. Moreover, oncogenic driver mutations have not been described for up to 30% of all melanomas. In order to obtain a more complete picture of the mutational landscape of melanoma, more recent studies used high-throughput DNA sequencing technologies. A number of new oncogene candidates such as MAPK1/2, ERBB4, GRIN2A, GRM3, RAC1, and PREX2 were identified. Their particular role in melanoma biology is currently under investigation. Evidence for the functional relevance of some of these new oncogene candidates has been provided in in vitro and in vivo experiments. However, these findings await further validation in clinical studies. This review provides an overview on well-known melanoma oncogenes and new oncogene candidates, based on recent high-throughput sequencing studies. The list of genes discussed herein is of course not complete but highlights some of the most significant of recent findings in this area. The new candidates may support more individualized treatment approaches for metastatic melanoma patients in the future.
Collapse
Affiliation(s)
- Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
28
|
Mace PD, Wallez Y, Egger MF, Dobaczewska MK, Robinson H, Pasquale EB, Riedl SJ. Structure of ERK2 bound to PEA-15 reveals a mechanism for rapid release of activated MAPK. Nat Commun 2013; 4:1681. [PMID: 23575685 PMCID: PMC3640864 DOI: 10.1038/ncomms2687] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 02/28/2013] [Indexed: 02/08/2023] Open
Abstract
ERK1/2 kinases are the principal effectors of a central signaling cascade that converts extracellular stimuli into cell proliferation and migration responses and, when deregulated, can promote cell oncogenic transformation. The scaffolding protein PEA-15 is a death effector domain (DED) protein that directly interacts with ERK1/2 and affects ERK1/2 subcellular localization and phosphorylation. Here, to understand this ERK1/2 signaling complex, we have solved the crystal structures of PEA-15 bound to three different ERK2 phospho-conformers. The structures reveal that PEA-15 uses a bipartite binding mode, occupying two key docking sites of ERK2. Remarkably, PEA-15 can efficiently bind the ERK2 activation loop in the critical Thr-X-Tyr region in different phosphorylation states. PEA-15 binding triggers an extended allosteric conduit in dually phosphorylated ERK2, disrupting key features of active ERK2. At the same time PEA-15 binding protects ERK2 from dephosphorylation, thus setting the stage for immediate ERK activity upon its release from the PEA-15 inhibitory complex.
Collapse
Affiliation(s)
- Peter D Mace
- Program in Apoptosis and Cell Death Research, Cancer Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Melanoma genotypes and phenotypes get personal. J Transl Med 2013; 93:858-67. [PMID: 23817084 DOI: 10.1038/labinvest.2013.84] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 05/28/2013] [Accepted: 06/01/2013] [Indexed: 01/23/2023] Open
Abstract
Traditionally, the diagnosis of metastatic melanoma was terminal to most patients. However, the advancements towards understanding the fundamental etiology, pathophysiology, and treatment have raised melanoma to the forefront of contemporary medicine. Indeed, the evidence of durable remissions are being heard ever more frequently in clinics around the globe. Despite having more gene mutations per cell than any other type of cancer, investigators are overcoming complex genomic landscapes, signaling pathways, and immune checkpoints by generating novel technological methods and clinical protocols with breath-taking speed. Significant progress in deciphering molecular genetics, epigenetics, kinase-driven networks, metabolomics, and immune-enhancing pathways to achieve personalized and positive outcomes has truly provided new hope for melanoma patients. However, obstacles requiring breakthroughs include understanding the influence of sunlight exposure on melanoma etiology, and overcoming all too frequently acquired drug resistance, complicating targeted therapy. Pathologists continue to have critically important roles in advancing the field, particularly in the area of transitioning from microscope-based diagnostic reports to pharmacogenomics through molecularly informed tumor boards. Although melanoma is no longer considered just 'one disease', pathologists will continue this rapidly progressing and exciting journey to identify tumor subtypes, to utilize tumorgraft or so-called patient-derived xenograft (PDX) models, and to develop companion diagnostics to keep pace with the bewildering breakthroughs occurring on a regular basis. Exactly which combination of drugs will ultimately be required to eradicate melanoma cells remains to be determined. However, it is clear that pathologists who are as dedicated to melanoma as the pioneering pathologist Dr Sidney Farber was committed to childhood cancers, will be required as the battle against melanoma continues. In this review, we describe what sets melanoma apart from other tumors, and demonstrate how lessons learned in the melanoma clinic are being transferred to many other types of aggressive neoplasms.
Collapse
|
30
|
Pópulo H, Tavares S, Faustino A, Nunes JB, Lopes JM, Soares P. GNAQ and BRAF mutations show differential activation of the mTOR pathway in human transformed cells. PeerJ 2013; 1:e104. [PMID: 23904987 PMCID: PMC3728761 DOI: 10.7717/peerj.104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 06/23/2013] [Indexed: 12/31/2022] Open
Abstract
Somatic mutations in GNAQ gene were described as being the main oncogenic activation in uveal melanomas, whereas mutations in BRAF gene have been described as a key genetic alteration that contributes to skin melanoma development. We have previously reported differential activation of the MAPK and AKT/mTOR signalling pathways in uveal and skin melanomas harbouring, respectively, GNAQ and BRAF mutations. The aim of this work was to compare the functional effect of GNAQ and BRAF mutations in mTOR and MAPK pathway activation, cell proliferation and apoptosis. In this work, we performed transient transfection of HEK293 cells with BRAFWT, BRAFV 600E, GNAQWT, GNAQQ209P and GNAQQ209L vectors. We treated melanoma cell lines displaying different BRAF and GNAQ mutational status with the mTOR inhibitor RAD001 and with the MEK1/2 inhibitor U0126 and evaluated the effects in the growth of the cell lines and in mTOR and MAPK pathway effectors expression. At variance with the significant increase in the level of pmTOR Ser2448 and pS6 Ser235/236 proteins observed in cells transfected with BRAF vectors, no significant alteration in mTOR pathway effectors was observed in cells transfected with the three GNAQ expressing vectors. Also, GNAQ overexpression enhances Stat3 activation, which might mediate GNAQ oncogenic effects. None of the vectors led to significant differences in proliferation or apoptosis in the transfected cell lines. Cell lines harbouring a BRAF mutation were more sensitive to RAD001 treatment. U0126 leads to the reduction of MAPK and mTOR pathways activation in all cell lines tested. Our results indicate that GNAQ and BRAF activation drive distinct intracellular signalling pathways that may be useful for therapeutic decisions in human melanomas.
Collapse
Affiliation(s)
- Helena Pópulo
- Institute of Molecular Pathology and Immunology, University of Porto , Porto , Portugal
| | | | | | | | | | | |
Collapse
|
31
|
Al-Mulla F, Bitar MS, Taqi Z, Yeung KC. RKIP: much more than Raf kinase inhibitory protein. J Cell Physiol 2013; 228:1688-702. [PMID: 23359513 DOI: 10.1002/jcp.24335] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/16/2013] [Indexed: 12/11/2022]
Abstract
From its discovery as a phosphatidylethanolamine-binding protein in bovine brain to its designation as a physiological inhibitor of Raf kinase protein, RKIP has emerged as a critical molecule for maintaining subdued, well-orchestrated cellular responses to stimuli. The disruption of RKIP in a wide range of pathologies, including cancer, Alzheimer's disease, and pancreatitis, makes it an exciting target for individualized therapy and disease-specific interventions. This review attempts to highlight recent advances in the RKIP field underscoring its potential role as a master modulator of many pivotal intracellular signaling cascades that control cellular growth, motility, apoptosis, genomic integrity, and therapeutic resistance. Specific biological and functional niches are highlighted to focus future research towards an enhanced understanding of the multiple roles of RKIP in health and disease.
Collapse
Affiliation(s)
- Fahd Al-Mulla
- Faculty of Medicine, Department of Pathology, Kuwait University Health Sciences Centre, Safat, Kuwait.
| | | | | | | |
Collapse
|
32
|
Long GV, Wilmott JS, Haydu LE, Tembe V, Sharma R, Rizos H, Thompson JF, Howle J, Scolyer RA, Kefford RF. Effects of BRAF inhibitors on human melanoma tissue before treatment, early during treatment, and on progression. Pigment Cell Melanoma Res 2013; 26:499-508. [PMID: 23557327 DOI: 10.1111/pcmr.12098] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 03/12/2013] [Indexed: 01/09/2023]
Abstract
Selective BRAF inhibitors (BRAFi) are a standard of care for the treatment of BRAF(V) (600) -mutant metastatic melanoma. We analyzed a unique set of serial triplicate human metastatic melanoma tumor biopsies to identify biomarkers of BRAFi response and resistance. Morphologic features and immunohistochemical biomarkers were analyzed in 37 metastatic melanoma biopsies at pretreatment (PRE), early during treatment (EDT), and on progression (PROG) from 15 patients treated with a BRAFi and correlated with response and outcome. At EDT, proliferative markers decreased regardless of response, whereas markers of cell death increased in responders. High expression of nuclear p27 at baseline was the strongest predictor of a poorer OS and predicted worse response. The results show that BRAFi are universally antiproliferative, regardless of clinical response, whereas markers of cell death increased only in responders. The addition of therapies targeting the cell cycle machinery may improve the response and duration of BRAFi, and investigation of the mechanisms of apoptosis may provide additional therapeutic targets.
Collapse
|
33
|
|
34
|
Smith KB, Tran LM, Tam BM, Shurell EM, Li Y, Braas D, Tap WD, Christofk HR, Dry SM, Eilber FC, Wu H. Novel dedifferentiated liposarcoma xenograft models reveal PTEN down-regulation as a malignant signature and response to PI3K pathway inhibition. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1400-11. [PMID: 23416162 DOI: 10.1016/j.ajpath.2013.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/14/2012] [Accepted: 01/03/2013] [Indexed: 10/27/2022]
Abstract
Liposarcoma is a type of soft tissue sarcoma that exhibits poor survival and a high recurrence rate. Treatment is generally limited to surgery and radiation, which emphasizes the need for better understanding of this disease. Because very few in vivo and in vitro models can reproducibly recapitulate the human disease, we generated several xenograft models from surgically resected human dedifferentiated liposarcoma. All xenografts recapitulated morphological and gene expression characteristics of the patient tumors after continuous in vivo passages. Importantly, xenograftability was directly correlated with disease-specific survival of liposarcoma patients. Thus, the ability for the tumor of a patient to engraft may help identify those patients who will benefit from more aggressive treatment regimens. Gene expression analyses highlighted the association between xenograftability and a unique gene expression signature, including down-regulated PTEN tumor-suppressor gene expression and a progenitor-like phenotype. When treated with the PI3K/AKT/mTOR pathway inhibitor rapamycin alone or in combination with the multikinase inhibitor sorafenib, all xenografts responded with increased lipid content and a more differentiated gene expression profile. These human xenograft models may facilitate liposarcoma research and accelerate the generation of readily translatable preclinical data that could ultimately influence patient care.
Collapse
Affiliation(s)
- Kathleen B Smith
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kim SW, Kim HK, Lee JI, Jang HW, Choe JH, Kim JH, Kim JS, Hur KY, Kim JH, Chung JH. ERK phosphorylation is not increased in papillary thyroid carcinomas with BRAF(V600E) mutation compared to that of corresponding normal thyroid tissues. Endocr Res 2013; 38:89-97. [PMID: 23544999 DOI: 10.3109/07435800.2012.723292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND An association between a BRAF(V600E) mutation and upregulation of mitogen-activated protein kinase (MAPK) pathways in human papillary thyroid carcinoma (PTC) tissues has not been demonstrated well outside of in vitro studies. The aims of this study were to evaluate the activation status of extracellular signal-regulated kinase 1/2 (ERK1/2) in human PTCs with BRAF(V600E) mutations compared to that of corresponding normal thyroid tissue and to determine the expressions of Raf kinase inhibitor protein (RKIP) and MAPK phosphatase 3 (MKP-3), possible regulators of ERK1/2 activation. METHODS We analyzed the presence of BRAF(V600E) mutation and the expressions of BRAF, total ERK, p-ERK, RKIP, and MKP-3 in 33 PTCs and corresponding normal thyroid gland tissues using western blot analysis. RESULTS BRAF(V600E) mutation was found in 28 (84.8%) of 33 PTCs, 96.4% (27/28) of which showed decreased p-ERK activity, while 75% (21/28) showed increased MKP-3 expression. There were significant differences in p-ERK and MKP-3 expressions between BRAF(V600E) (+) PTCs and normal thyroid glands (p < 0.001). There were no differences in expressions of BRAF, total ERK, and RKIP between PTCs and normal thyroid tissue, irrespective of the presence of BRAF(V600E) mutation. CONCLUSIONS In human BRAF(V600E) (+) PTCs, ERK phosphorylation is decreased compared to normal thyroid glands and the observed decrease in ERK1/2 MAPK phosphorylation in BRAF(V600E) (+) PTCs may be associated with increased MKP-3 activity.
Collapse
Affiliation(s)
- Sun Wook Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Pathria G, Wagner C, Wagner SN. Inhibition of CRM1-mediated nucleocytoplasmic transport: triggering human melanoma cell apoptosis by perturbing multiple cellular pathways. J Invest Dermatol 2012; 132:2780-90. [PMID: 22832492 DOI: 10.1038/jid.2012.233] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Development of multiple drug resistance mechanisms in melanomas necessitates the identification of new drug targets, which when inhibited could impact multiple cellular pathways, thus circumventing potential resistance. By performing complementary DNA microarray analysis, we identified four key components of the nucleocytoplasmic transport machinery-CRM1, RAN (RAN-GTPase), RANGAP1, and RANBP1-to be overexpressed in human melanoma metastases. Chromosome region maintenance 1 (CRM1) inhibition induced a marked depletion of prosurvival/cytoplasmic extracellular signal-regulated kinase 1/2 (Erk1/2) and p90 ribosomal S6 kinase1 and elicited persistent Erk-signaling hyperactivation. Consistently, CRM1 inhibition inflicted extensive apoptosis in melanoma cells while sparing nontransformed melanocytes and primary lung fibroblasts. Apoptosis required both the intrinsic and extrinsic apoptotic pathways and was associated with a nuclear entrapment and downregulation of the antiapoptotic CRM1 target protein, Survivin. Apoptosis was preceded by a G1 cell-cycle arrest, and even though CRM1 inhibition mediated marked p53 and p21 induction in wild-type p53 melanoma cells, the latter's silencing or inactivation failed to alleviate apoptosis. Notably, CRM1 inhibition induced cell line-specific, G1 to S progression-retarding changes in the expression of multiple cell-cycle regulatory proteins, thus potentially explaining p53 dispensability. We propose CRM1 as a potential therapeutic target in human melanoma, whose inhibition induces loss of prosurvival/cytoplasmic Erk1/2, mediates persistent Erk hyperactivation, and initiates a multitude of cell context-dependent molecular events to trigger G1 arrest followed by massive apoptosis.
Collapse
Affiliation(s)
- Gaurav Pathria
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| | | | | |
Collapse
|
37
|
|
38
|
Hunter SM, Gorringe KL, Christie M, Rowley SM, Bowtell DD, Campbell IG. Pre-Invasive Ovarian Mucinous Tumors Are Characterized by CDKN2A and RAS Pathway Aberrations. Clin Cancer Res 2012; 18:5267-77. [DOI: 10.1158/1078-0432.ccr-12-1103] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
39
|
Schoenewolf NL, Bull C, Belloni B, Holzmann D, Tonolla S, Lang R, Mihic-Probst D, Andres C, Dummer R. Sinonasal, genital and acrolentiginous melanomas show distinct characteristics of KIT expression and mutations. Eur J Cancer 2012; 48:1842-52. [DOI: 10.1016/j.ejca.2012.02.049] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 02/07/2012] [Accepted: 02/15/2012] [Indexed: 11/25/2022]
|
40
|
Colombino M, Capone M, Lissia A, Cossu A, Rubino C, De Giorgi V, Massi D, Fonsatti E, Staibano S, Nappi O, Pagani E, Casula M, Manca A, Sini M, Franco R, Botti G, Caracò C, Mozzillo N, Ascierto PA, Palmieri G. BRAF/NRAS Mutation Frequencies Among Primary Tumors and Metastases in Patients With Melanoma. J Clin Oncol 2012; 30:2522-9. [DOI: 10.1200/jco.2011.41.2452] [Citation(s) in RCA: 359] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Purpose The prevalence of BRAF, NRAS, and p16CDKN2A mutations during melanoma progression remains inconclusive. We investigated the prevalence and distribution of mutations in these genes in different melanoma tissues. Patients and Methods In all, 291 tumor tissues from 132 patients with melanoma were screened. Paired samples of primary melanomas (n = 102) and synchronous or asynchronous metastases from the same patients (n = 165) were included. Tissue samples underwent mutation analysis (automated DNA sequencing). Secondary lesions included lymph nodes (n = 84), and skin (n = 36), visceral (n = 25), and brain (n = 44) sites. Results BRAF/NRAS mutations were identified in 58% of primary melanomas (43% BRAF; 15% NRAS); 62% in lymph nodes, 61% subcutaneous, 56% visceral, and 70% in brain sites. Mutations were observed in 63% of metastases (48% BRAF; 15% NRAS), a nonsignificant increase in mutation frequency after progression from primary melanoma. Of the paired samples, lymph nodes (93% consistency) and visceral metastases (96% consistency) presented a highly similar distribution of BRAF/NRAS mutations versus primary melanomas, with a significantly less consistent pattern in brain (80%) and skin metastases (75%). This suggests that independent subclones are generated in some patients. p16CDKN2A mutations were identified in 7% and 14% of primary melanomas and metastases, with a low consistency (31%) between secondary and primary tumor samples. Conclusion In the era of targeted therapies, assessment of the spectrum and distribution of alterations in molecular targets among patients with melanoma is needed. Our findings about the prevalence of BRAF/NRAS/p16CDKN2A mutations in paired tumor lesions from patients with melanoma may be useful in the management of this disease.
Collapse
Affiliation(s)
- Maria Colombino
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Mariaelena Capone
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Amelia Lissia
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Antonio Cossu
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Corrado Rubino
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Vincenzo De Giorgi
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Daniela Massi
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Ester Fonsatti
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Stefania Staibano
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Oscar Nappi
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Elena Pagani
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Milena Casula
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Antonella Manca
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - MariaCristina Sini
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Renato Franco
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Gerardo Botti
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Corrado Caracò
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Nicola Mozzillo
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Paolo A. Ascierto
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| | - Giuseppe Palmieri
- Maria Colombino, Milena Casula, Antonella Manca, MariaCristina Sini, and Giuseppe Palmieri, Istituto Chimica Biomolecolare, Consiglio Nazionale delle Ricerche; Amelia Lissia and Antonio Cossu, Azienda Ospedaliero Universitaria; Corrado Rubino, Università di Sassari, Sassari; Mariaelena Capone, Renato Franco, Gerardo Botti, Corrado Caracò, Nicola Mozzillo, and Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione Pascale; Stefania Staibano, Università Federico II di Napoli; Oscar Nappi, Ospedale
| |
Collapse
|
41
|
Kim JE, Stones C, Joseph WR, Leung E, Finlay GJ, Shelling AN, Phillips WA, Shepherd PR, Baguley BC. Comparison of growth factor signalling pathway utilisation in cultured normal melanocytes and melanoma cell lines. BMC Cancer 2012; 12:141. [PMID: 22475322 PMCID: PMC3352269 DOI: 10.1186/1471-2407-12-141] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 04/04/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The phosphatidylinositol-3-kinase (PI3K-PKB), mitogen activated protein kinase (MEK-ERK) and the mammalian target of rapamycin (mTOR- p70S6K), are thought to regulate many aspects of tumour cell proliferation and survival. We have examined the utilisation of these three signalling pathways in a number of cell lines derived from patients with metastatic malignant melanoma of known PIK3CA, PTEN, NRAS and BRAF mutational status. METHODS Western blotting was used to compare the phosphorylation status of components of the PI3K-PKB, MEK-ERK and mTOR-p70S6K signalling pathways, as indices of pathway utilisation. RESULTS Normal melanocytes could not be distinguished from melanoma cells on the basis of pathway utilisation when grown in the presence of serum, but could be distinguished upon serum starvation, where signalling protein phosphorylation was generally abrogated. Surprisingly, the differential utilisation of individual pathways was not consistently associated with the presence of an oncogenic or tumour suppressor mutation of genes in these pathways. CONCLUSION Utilisation of the PI3K-PKB, MEK-ERK and mTOR-p70S6K signalling pathways in melanoma, as determined by phosphorylation of signalling components, varies widely across a series of cell lines, and does not directly reflect mutation of genes coding these components. The main difference between cultured normal melanocytes and melanoma cells is not the pathway utilisation itself, but rather in the serum dependence of pathway utilisation.
Collapse
Affiliation(s)
- Ji Eun Kim
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
- Auckland Cancer Society Research Centre, Private Bag 92019, The University of Auckland, Auckland, New Zealand
| | - Clare Stones
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Wayne R Joseph
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Euphemia Leung
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Graeme J Finlay
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Andrew N Shelling
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
| | - Wayne A Phillips
- Department of Surgery, Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre and University of Melbourne, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Bruce C Baguley
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
42
|
Abstract
RAF kinase inhibitor protein (RKIP) is a negative regulator of the RAS-mitogen-activated protein kinase/extracellular signal-regulated kinase signaling cascade. We investigated its role in acute myeloid leukemia (AML), an aggressive malignancy arising from hematopoietic stem and progenitor cells (HSPCs). Western blot analysis revealed loss of RKIP expression in 19/103 (18%) primary AML samples and 4/17 (24%) AML cell lines but not in 10 CD34+ HSPC specimens. In in-vitro experiments with myeloid cell lines, RKIP overexpression inhibited cellular proliferation and colony formation in soft agar. Analysis of two cohorts with 103 and 285 AML patients, respectively, established a correlation of decreased RKIP expression with monocytic phenotypes. RKIP loss was associated with RAS mutations and in transformation assays, RKIP decreased the oncogenic potential of mutant RAS. Loss of RKIP further related to a significantly longer relapse-free survival and overall survival in uni- and multivariate analyses. Our data show that RKIP is frequently lost in AML and correlates with monocytic phenotypes and mutations in RAS. RKIP inhibits proliferation and transformation of myeloid cells and decreases transformation induced by mutant RAS. Finally, loss of RKIP seems to be a favorable prognostic parameter in patients with AML.
Collapse
|
43
|
Zhao YB, Wang YH, Abuduwaili•Wushour. Application of SELDI-TOF-MS and LCM to screen protein markers for early diagnosis of liver metastasis of colorectal carcinoma. Shijie Huaren Xiaohua Zazhi 2012; 20:296-303. [DOI: 10.11569/wcjd.v20.i4.296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To screen protein markers for liver metastases of colorectal cancer using surface-enhanced laser desorption/ionization time-of-flight mass spectrometry protein chip (SELDI-TOF-MS) in combination with laser capture microdissection (LCM) technology.
METHODS: Normal colorectal cells, primary tumor cells and liver metastatic cells were obtained from 24 colorectal cancer patients with liver metastases using the LCM technology. Protein profiling was performed using SELDI-TOF-MS technology. Differentially expressed proteins were analyzed using Biomarker Wizard software and identified by querying the database.
RESULTS: Fifteen differentially expressed proteins were found between primary tumor cells and normal colorectal cells, of which 12 were up-regulated and 3 down-regulated. Nine differentially expressed proteins were found between liver metastatic cells and primary tumor cells, of which 5 were up-regulated and 4 down-regulated. Twenty differentially expressed proteins were identified by querying ExPasy protein database, including integral membrane protein 2C, DNA repair protein RAD51 homolog 4, cell cycle checkpoint protein RAD1, human epididymis protein 4, centromere protein R, and pleckstrin homology domain family member 3. Apoptosis regulator Bax-γ, S100A11, Raf kinase inhibitor protein (RKIP) and heat shock protein 27 (HSP-27) displayed most obvious differential expression among normal colorectal cells, primary tumor cells and liver metastatic cells (all P < 0. 01).
CONCLUSION: SELDI-TOF-MS technology combined with LCM may allow to screen highly sensitive and specific protein markers for colorectal cancer and liver metastases of colorectal cancer.
Collapse
|
44
|
Tyr phosphatase-mediated P-ERK inhibition suppresses senescence in EIA + v-raf transformed cells, which, paradoxically, are apoptosis-protected in a MEK-dependent manner. Neoplasia 2011; 13:120-30. [PMID: 21403838 DOI: 10.1593/neo.101152] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 11/12/2010] [Accepted: 11/18/2010] [Indexed: 01/01/2023] Open
Abstract
Activation of the Ras-Raf-extracellular signal-regulated kinase (ERK) pathway causes not only proliferation and suppression of apoptosis but also the antioncogenic response of senescence. How these contrasting effects are reconciled to achieve cell transformation and cancer formation is poorly understood. In a system of two-step carcinogenesis (dedifferentiated PC EIA, transformed PC EIA-polyoma-middle T [PC EIA + Py] and PC EIA-v-raf [PC EIA + raf] cells], v-raf cooperated with EIA by virtue of a strong prosurvival effect, not elicited by Py-middle T, evident toward serum-deprivation-and H(2)O(2)-induced apoptosis. Apoptosis was detected by DNA fragmentation and annexin V staining. The prosurvival function of v-raf was, in part, mitogen-activated protein kinase/ERK kinase (MEK)-dependent, as shown by pharmacological MEK inhibition. The MEK-dependent antiapoptotic effect of v-raf was exerted despite a lower level of P-ERK1/2 in EIA + raf cells with respect to EIA + Py/EIA cells, which was dependent on a high tyrosine phosphatase activity, as shown by orthovanadate blockade. An ERK1/2 tyrosine phosphatase was likely involved. The high tyrosine phosphatase activity was instrumental to the complete suppression of senescence, detected by β-galactosidase activity, because tyrosine phosphatase blockade induced senescence in EIA + raf but not in EIA + Py cells. High tyrosine phosphatase activity and evasion from senescence were confirmed in an anaplastic thyroid cancer cell line. Therefore, besides EIA, EIA + raf cells suppress senescence through a new mechanism, namely, phosphatase-mediated P-ERK1/2 inhibition, but, paradoxically, retain the oncogenic effects of the Raf-ERK pathway. We propose that the survival effect of Raf is not a function of absolute P-ERK1/2 levels at a given time but is rather dynamically dependent on greater variations after an apoptotic stimulus.
Collapse
|
45
|
Chen H, Takahara M, Oba J, Xie L, Chiba T, Takeuchi S, Tu Y, Nakahara T, Uchi H, Moroi Y, Furue M. Clinicopathologic and prognostic significance of SATB1 in cutaneous malignant melanoma. J Dermatol Sci 2011; 64:39-44. [PMID: 21767935 DOI: 10.1016/j.jdermsci.2011.06.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 05/29/2011] [Accepted: 06/15/2011] [Indexed: 01/06/2023]
Abstract
BACKGROUND Special AT-rich sequence-binding protein-1 (SATB1), a new type of gene regulator, has been reported to be expressed in several human cancers and may have malignant potential. However, no data on SATB1 expression and its relationship to tumor progression in cutaneous malignant melanoma (CMM) has yet been reported. OBJECTIVE We examined the immunohistochemical expression of SATB1 in CMM to determine whether it could serve as a prognostic marker. METHODS A total of 97 samples of primary CMM and controls were immunostained for SATB1. The following clinicopathologic variables were evaluated: age, gender, subtype, SATB1 expression, Breslow thickness, Clark level, presence of ulceration, lymph node metastasis, distant metastasis, and survival. Statistical analyses were performed to assess for associations. Several parameters were analyzed for survival using the Kaplan-Meier method and Cox proportional-hazards model. RESULTS Forty cases (85.1%) of CMM showed positive staining for SATB1 by immunohistochemistry. The intensity of SATB1 staining was significantly higher in CMM than in nevus NV and normal skin (NS) (P < 0.01). High SATB1 expression was significantly correlated with Breslow thickness, Clark level, mortality, presence of ulceration, and lymph node metastasis (P < 0.01). Moreover, Kaplan-Meier analysis revealed that SATB1 overexpression was significantly associated with worse survival (P < 0.01). Further univariate analysis and multivariate regression analysis indicated that SATB1 expression was an independent prognostic marker for CMM (P = 0.03). CONCLUSIONS The overexpression of SATB1 correlated with metastatic potential of CMM and is a novel independent prognostic marker for predicting outcome.
Collapse
Affiliation(s)
- Hongxiang Chen
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Huerta-Yepez S, Yoon NK, Hernandez-Cueto A, Mah V, Rivera-Pazos CM, Chatterjee D, Vega MI, Maresh EL, Horvath S, Chia D, Bonavida B, Goodglick L. Expression of phosphorylated raf kinase inhibitor protein (pRKIP) is a predictor of lung cancer survival. BMC Cancer 2011; 11:259. [PMID: 21689459 PMCID: PMC3134426 DOI: 10.1186/1471-2407-11-259] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 06/21/2011] [Indexed: 12/03/2022] Open
Abstract
Background Raf-1 kinase inhibitor protein (RKIP) has been reported to negatively regulate signal kinases of major survival pathways. RKIP activity is modulated in part by phosphorylation on Serine 153 by protein kinase C, which leads to dissociation of RKIP from Raf-1. RKIP expression is low in many human cancers and represents an indicator of poor prognosis and/or induction of metastasis. The prognostic power has typically been based on total RKIP expression and has not considered the significance of phospho-RKIP. Methods The present study examined the expression levels of both RKIP and phospho-RKIP in human lung cancer tissue microarray proteomics technology. Results Total RKIP and phospho-RKIP expression levels were similar in normal and cancerous tissues. phospho-RKIP levels slightly decreased in metastatic lesions. However, the expression levels of phospho-RKIP, in contrast to total RKIP, displayed significant predictive power for outcome with normal expression of phospho-RKIP predicting a more favorable survival compared to lower levels (P = 0.0118); this was even more pronounced in more senior individuals and in those with early stage lung cancer. Conclusions This study examines for the first time, the expression profile of RKIP and phospho-RKIP in lung cancer. Significantly, we found that phospho-RKIP was a predictive indicator of survival.
Collapse
Affiliation(s)
- Sara Huerta-Yepez
- Unidad de Investigacion en Enferemedades Oncologicas, Hospital Infantil de Mexico, Federico Gomez, SSa, Mexico
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Puzanov I, Burnett P, Flaherty KT. Biological challenges of BRAF inhibitor therapy. Mol Oncol 2011; 5:116-23. [PMID: 21393075 DOI: 10.1016/j.molonc.2011.01.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 01/27/2011] [Indexed: 12/19/2022] Open
Abstract
Activating mutations in BRAF, a constituent of the map kinase pathway, were first discovered as being most prevalent in melanoma in 2002. Only recently have potent and selective, orally available inhibitors of BRAF emerged for clinical testing and demonstrated clear evidence of tumor regression in the majority of patients whose tumors harbor a BRAF mutation. While these early observations suggest that the BRAF targeted therapy will become part of the standard treatment paradigm for patients with advanced melanoma, it is also clear that a majority of these responses are incomplete and temporary. Therefore, the focus of the melanoma field has shifted to understanding the limits of the first generation of selective BRAF inhibitors with regard to safety and efficacy, the context of somatic genetic changes that accompany BRAF, and the combination regimens that target distinct elements of melanoma pathophysiology.
Collapse
Affiliation(s)
- Igor Puzanov
- Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | | | | |
Collapse
|
48
|
Polyclonality of BRAF mutations in primary melanoma and the selection of mutant alleles during progression. Br J Cancer 2011; 104:464-8. [PMID: 21224857 PMCID: PMC3049568 DOI: 10.1038/sj.bjc.6606072] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Oncogenic BRAF mutation had been considered to be a founder event in the formation of melanocytic tumours; however, we recently argued against this notion by showing marked polyclonality of BRAF mutations in acquired melanocytic nevi (Lin et al, J Natl Cancer Inst., 2009; 101:1423-7). Here, we tested whether similar heterogeneity of BRAF mutations exists in primary melanomas. METHODS We isolated and sequenced single melanoma cells from five primary melanoma tissues using antibodies against human high-molecular-weight melanoma-associated antigen. We also examined 10 primary melanomas by the sensitive Mutector assay detecting the BRAF(V600E) mutation, as well as by cloning and sequencing of separated alleles. Furthermore, we estimated the frequency of BRAF mutant alleles in paired samples of primary tumour and recurrence or metastasis in three patients. RESULTS Single-cell mutation analyses revealed that four of five primary melanomas contained both BRAF-wild-type and BRAF-mutant tumour cells. Tumour heterogeneity in terms of BRAF mutations was also shown in 8 of 10 primary melanomas. Selection of BRAF mutant alleles during progression was demonstrated in all the three patients. CONCLUSION Acquisition of a BRAF mutation is not a founder event, but may be one of the multiple clonal events in melanoma development, which is selected for during the progression.
Collapse
|
49
|
Nakamura S, Yagyu T, Takemura T, Tan L, Nagata Y, Yokota D, Hirano I, Shibata K, Fujie M, Fujisawa S, Ohnishi K. Bcr-Abl-mediated Raf kinase inhibitor protein suppression promotes chronic myeloid leukemia progenitor cells proliferation. ACTA ACUST UNITED AC 2011. [DOI: 10.4236/scd.2011.13006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
50
|
Flaherty KT, McArthur G. BRAF, a target in melanoma: implications for solid tumor drug development. Cancer 2010; 116:4902-13. [PMID: 20629085 DOI: 10.1002/cncr.25261] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The successful translation of therapies targeting signal-transduction pathways that are activated by oncogenes has provided a model for molecularly targeted therapy, and the identification of mutations in v-raf murine sarcoma viral oncogene homolog B1 (BRAF), a serine/threonine kinase, has turned the attention of the melanoma field toward this concept. The current review indicated that BRAF represents an important target in cancer, in part because it is present in 7% of all cancers and also because it represents the first intracellular signaling molecule that is activated by point mutations for which single-agent therapy appears to have efficacy. Therapy for advanced melanoma has progressed slowly over the past 3 decades, although significant advances have been made in other cancers with the application of cytotoxic chemotherapy and targeted therapies. However, in melanoma, cytotoxic chemotherapies have severe limits, chemotherapy does not convincingly improve on the natural history of metastatic disease and has no role in the adjuvant setting, and cytokine therapy may have a niche in both the adjuvant and metastatic settings but confers only a modest benefit to a small proportion of patients at the cost of severe toxicity. Thus, there are few other cancers in which completely novel therapies are so highly prioritized in clinical research. Understanding network of signal-transduction pathways and how that network may adapt to BRAF inhibition or mitogen-activated protein kinase kinase inhibition will point to the next generation of clinical trials investigating rational combination regimens. The current investigations in melanoma will create a set of hypotheses to be tested in each cancer that harbors BRAF mutations.
Collapse
Affiliation(s)
- Keith T Flaherty
- Division of Hematology/Oncology, Massuchusetts General Hospital Cancer Center, Boston, Massachusetts, USA.
| | | |
Collapse
|