1
|
Mousa DPV, Mavrovounis G, Argyropoulos D, Stranjalis G, Kalamatianos T. Anaplastic Lymphoma Kinase (ALK) in Posterior Cranial Fossa Tumors: A Scoping Review of Diagnostic, Prognostic, and Therapeutic Perspectives. Cancers (Basel) 2024; 16:650. [PMID: 38339401 PMCID: PMC10854950 DOI: 10.3390/cancers16030650] [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: 12/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Anaplastic Lymphoma Kinase (ALK) has been implicated in several human cancers. This review aims at mapping the available literature on the involvement of ALK in non-glial tumors localized in the posterior cranial fossa and at identifying diagnostic, prognostic, and therapeutic considerations. Following the PRISMA-ScR guidelines, studies were included if they investigated ALK's role in primary CNS, non-glial tumors located in the posterior cranial fossa. A total of 210 manuscripts were selected for full-text review and 16 finally met the inclusion criteria. The review included 55 cases of primary, intracranial neoplasms with ALK genetic alterations and/or protein expression, located in the posterior fossa, comprising of medulloblastoma, anaplastic large-cell lymphoma, histiocytosis, inflammatory myofibroblastic tumors, and intracranial myxoid mesenchymal tumors. ALK pathology was investigated via immunohistochemistry or genetic analysis. Several studies provided evidence for potential diagnostic and prognostic value for ALK assessment as well as therapeutic efficacy in its targeting. The available findings on ALK in posterior fossa tumors are limited. Nevertheless, previous findings suggest that ALK assessment is of diagnostic and prognostic value in medulloblastoma (WNT-activated). Interestingly, a substantial proportion of ALK-positive/altered CNS histiocytoses thus far identified have been localized in the posterior fossa. The therapeutic potential of ALK inhibition in histiocytosis warrants further investigation.
Collapse
Affiliation(s)
| | - Georgios Mavrovounis
- Department of Neurosurgery, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41334 Larissa, Greece;
- Department of Neurosurgery, Evangelismos Hospital, School of Medicine, Faculty of Health Sciences, National and Kapodistrian University of Athens, 10676 Athens, Greece;
| | - Dionysios Argyropoulos
- Department of Psychiatry, Eginition Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - George Stranjalis
- Department of Neurosurgery, Evangelismos Hospital, School of Medicine, Faculty of Health Sciences, National and Kapodistrian University of Athens, 10676 Athens, Greece;
| | - Theodosis Kalamatianos
- Department of Neurosurgery, Evangelismos Hospital, School of Medicine, Faculty of Health Sciences, National and Kapodistrian University of Athens, 10676 Athens, Greece;
| |
Collapse
|
2
|
Blandin AF, Giglio R, Graham MS, Garcia G, Malinowski S, Woods JK, Ramkissoon S, Ramkissoon L, Dubois F, Schoolcraft K, Tsai J, Wang D, Jones R, Vogelzang J, Pelton K, Becker S, Watkinson F, Sinai C, Cohen EF, Booker MA, Tolstorukov MY, Haemels V, Goumnerova L, Wright K, Kieran M, Fehnel K, Reardon D, Tauziede-Espariat A, Lulla R, Carcamo B, Chaleff S, Charest A, DeSmet F, Ligon AH, Dubuc A, Pages M, Varlet P, Wen PY, Alexander BM, Chi S, Alexandrescu S, Kittler R, Bachoo R, Bandopadhayay P, Beroukhim R, Ligon KL. ALK Amplification and Rearrangements Are Recurrent Targetable Events in Congenital and Adult Glioblastoma. Clin Cancer Res 2023; 29:2651-2667. [PMID: 36780194 PMCID: PMC10363218 DOI: 10.1158/1078-0432.ccr-21-3521] [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: 09/29/2021] [Revised: 10/31/2022] [Accepted: 02/07/2023] [Indexed: 02/14/2023]
Abstract
PURPOSE Anaplastic lymphoma kinase (ALK) aberrations have been identified in pediatric-type infant gliomas, but their occurrence across age groups, functional effects, and treatment response has not been broadly established. EXPERIMENTAL DESIGN We performed a comprehensive analysis of ALK expression and genomic aberrations in both newly generated and retrospective data from 371 glioblastomas (156 adult, 205 infant/pediatric, and 10 congenital) with in vitro and in vivo validation of aberrations. RESULTS ALK aberrations at the protein or genomic level were detected in 12% of gliomas (45/371) in a wide age range (0-80 years). Recurrent as well as novel ALK fusions (LRRFIP1-ALK, DCTN1-ALK, PRKD3-ALK) were present in 50% (5/10) of congenital/infant, 1.4% (3/205) of pediatric, and 1.9% (3/156) of adult GBMs. ALK fusions were present as the only candidate driver in congenital/infant GBMs and were sometimes focally amplified. In contrast, adult ALK fusions co-occurred with other oncogenic drivers. No activating ALK mutations were identified in any age group. Novel and recurrent ALK rearrangements promoted STAT3 and ERK1/2 pathways and transformation in vitro and in vivo. ALK-fused GBM cellular and mouse models were responsive to ALK inhibitors, including in patient cells derived from a congenital GBM. Relevant to the treatment of infant gliomas, we showed that ALK protein appears minimally expressed in the forebrain at perinatal stages, and no gross effects on perinatal brain development were seen in pregnant mice treated with the ALK inhibitor ceritinib. CONCLUSIONS These findings support use of brain-penetrant ALK inhibitors in clinical trials across infant, pediatric, and adult GBMs. See related commentary by Mack and Bertrand, p. 2567.
Collapse
Affiliation(s)
- Anne-Florence Blandin
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad institute of Harvard and MIT, Cambridge, MA, USA
| | - Ross Giglio
- Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | - Jared K. Woods
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad institute of Harvard and MIT, Cambridge, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | | | | | - Frank Dubois
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Jessica Tsai
- Dana-Farber Cancer Institute, Boston, MA, USA
- Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
| | - Dayle Wang
- Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | | | | | | | - Elizabeth F Cohen
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Matthew A Booker
- Department of Informatics and Analytics, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Veerle Haemels
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | | | - Karen Wright
- Dana-Farber Cancer Institute, Boston, MA, USA
- Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
| | - Mark Kieran
- Day One Biopharmaceuticals, Brisbane, CA 94005
| | - Katie Fehnel
- Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
| | | | | | - Rishi Lulla
- Hasbro Children's Hospital, Providence, RI, USA
| | - Benjamin Carcamo
- Texas Tech University, Health Science Center, Paul L. Foster School of Medicine, El Paso, TX, USA
- El Paso Children's Hospital, El Paso, TX, USA
| | | | - Alain Charest
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Frederik DeSmet
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Azra H. Ligon
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
- Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
| | - Adrian Dubuc
- Dana-Farber Cancer Institute, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | - Melanie Pages
- Department of Genetics, Institute Curie, Paris, France. INSERM U830, Laboratory of Translational Research in Pediatric Oncology, SIREDO Pediatric Oncology Center, Institute Curie, Paris, France
| | | | - Patrick Y. Wen
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Brian M. Alexander
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | - Susan Chi
- Dana-Farber Cancer Institute, Boston, MA, USA
- Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
| | - Sanda Alexandrescu
- Dana-Farber Cancer Institute, Boston, MA, USA
- Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
| | - Ralf Kittler
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Robert Bachoo
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pratiti Bandopadhayay
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad institute of Harvard and MIT, Cambridge, MA, USA
- Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
| | - Rameen Beroukhim
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad institute of Harvard and MIT, Cambridge, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | - Keith L. Ligon
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad institute of Harvard and MIT, Cambridge, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
- Boston Children's Cancer and Blood Disorder Center, Boston, MA, USA
- Dana-Farber Cancer Institute, Center for Patient Derived Models (CPDM), Boston, MA, USA
| |
Collapse
|
3
|
Azab MA. Expression of Anaplastic Lymphoma Kinase (ALK) in glioma and possible clinical correlations. A retrospective institutional study. Cancer Treat Res Commun 2023; 36:100703. [PMID: 37271069 DOI: 10.1016/j.ctarc.2023.100703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/04/2023] [Accepted: 03/31/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND Glioblastoma is considered the most aggressive primary brain tumor. Recurrence after treatment is a significant problem with a failed response to optimal treatment. The recurrence of GBM is linked to different cellular and molecular pathways. Nationwide, in Egypt, astrocytic tumors are the most commonly diagnosed CNS tumor. Anaplastic Lymphoma Kinase (ALK CD246) is an enzymatic protein (RTK) belonging to the insulin receptors superfamily. METHODS This is a retrospective study including sixty cases of astrocytic tumors (males = 40, mean age = 31.5), (females = 20, mean age = 37.77) obtained through collecting archived paraffin blocks of astrocytic tumor from the Pathology Department, Cairo University Faculty of Medicine during the period from January 2015 till January 2019. All cases were evaluated for ALK expression trying to find any clinical correlations with the clinical data. RESULTS Correlations were made using a scatterplot matrix correlogram. There was a significant correlation between tumor recurrence and ALK expression (r = 0.8, P < 0.01), and incidence of postoperative seizures (r = 0.8, P < 0.05), and between mean age and score tumor (r = 0.8, P < 0.05). CONCLUSION Expression of ALK was found to be abundant among high-grade gliomas and tumor recurrence rate was higher in ALK-positive patients. Further studies are needed to evaluate the potential use of ALK as a prognostic marker in cases of GBM.
Collapse
Affiliation(s)
- Mohammed A Azab
- Department of Neurosurgery, Cairo University Faculty of Medicine, Cairo, Egypt.
| |
Collapse
|
4
|
Pearce J, Khabra K, Nanji H, Stone J, Powell K, Martin D, Zebian B, Hettige S, Reisz Z, Bodi I, Al-Sarraj S, Bridges LR, Clarke M, Jones C, Mandeville HC, Vaidya S, Marshall LV, Carceller F. High grade gliomas in young children: The South Thames Neuro-Oncology unit experience and recent advances in molecular biology and targeted therapies. Pediatr Hematol Oncol 2021; 38:707-721. [PMID: 33900873 DOI: 10.1080/08880018.2021.1907493] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/04/2023]
Abstract
High grade gliomas (HGG) have a dismal prognosis with survival rates of 15-35%. Approximately 10-12% of pediatric HGG occur in young children and their molecular biology and clinical outcomes differ from those arising at older ages. We report on four children aged <5 years newly diagnosed with non-brainstem HGG between 2011 and 2018 who were treated with surgery and BBSFOP chemotherapy. Two died of tumor progression. The other two are still alive without radiotherapy at 3.8 and 3.9 years from diagnosis: one of whom remains disease-free off treatment; and the other one, whose tumor harbored a KCTD16:NTRK2 fusion, went on to receive larotrectinib. Additionally we review the general management, outcomes and latest updates in molecular biology and targeted therapies for young children with HGG. Infant gliomas can be stratified in molecular subgroups with clinically actionable oncogenic drivers. Chemotherapy-based strategies can avoid or delay the need for radiotherapy in young children with HGG. Harnessing the potential of NTRK, ALK, ROS1 and MET inhibitors offers the opportunity to optimize the therapeutic armamentarium to improve current outcomes for these children.
Collapse
Affiliation(s)
- Janice Pearce
- Children & Young People's Unit, Pediatric & Adolescent Neuro-Oncology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Komel Khabra
- Statistics Department, The Royal Marsden NHS Foundation Trust, London, UK
| | - Henry Nanji
- Statistics Department, The Royal Marsden NHS Foundation Trust, London, UK
| | - Joanna Stone
- Children & Young People's Unit, Pediatric & Adolescent Neuro-Oncology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Karen Powell
- Children & Young People's Unit, Pediatric & Adolescent Neuro-Oncology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Danielle Martin
- Children & Young People's Unit, Pediatric & Adolescent Neuro-Oncology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Bassel Zebian
- Neurosurgery Department, King's College Hospital NHS Foundation Trust, London, UK
| | - Samantha Hettige
- Neurosurgery Department, St George's Hospital NHS Foundation Trust, London, UK
| | - Zita Reisz
- Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, London, UK
| | - Istvan Bodi
- Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, London, UK
| | - Safa Al-Sarraj
- Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, London, UK
| | - Leslie R Bridges
- Department of Cellular Pathology, St George's Hospital NHS Foundation Trust, London, UK
| | - Matthew Clarke
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Chris Jones
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Henry C Mandeville
- Department of Radiation Oncology, The Royal Marsden NHS Foundation Trust, London, UK
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Sucheta Vaidya
- Children & Young People's Unit, Pediatric & Adolescent Neuro-Oncology, The Royal Marsden NHS Foundation Trust, London, UK
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Lynley V Marshall
- Children & Young People's Unit, Pediatric & Adolescent Neuro-Oncology, The Royal Marsden NHS Foundation Trust, London, UK
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Fernando Carceller
- Children & Young People's Unit, Pediatric & Adolescent Neuro-Oncology, The Royal Marsden NHS Foundation Trust, London, UK
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| |
Collapse
|
5
|
Defining Pathological Activities of ALK in Neuroblastoma, a Neural Crest-Derived Cancer. Int J Mol Sci 2021; 22:ijms222111718. [PMID: 34769149 PMCID: PMC8584162 DOI: 10.3390/ijms222111718] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroblastoma is a common extracranial solid tumour of childhood, responsible for 15% of cancer-related deaths in children. Prognoses vary from spontaneous remission to aggressive disease with extensive metastases, where treatment is challenging. Tumours are thought to arise from sympathoadrenal progenitor cells, which derive from an embryonic cell population called neural crest cells that give rise to diverse cell types, such as facial bone and cartilage, pigmented cells, and neurons. Tumours are found associated with mature derivatives of neural crest, such as the adrenal medulla or paraspinal ganglia. Sympathoadrenal progenitor cells express anaplastic lymphoma kinase (ALK), which encodes a tyrosine kinase receptor that is the most frequently mutated gene in neuroblastoma. Activating mutations in the kinase domain are common in both sporadic and familial cases. The oncogenic role of ALK has been extensively studied, but little is known about its physiological role. Recent studies have implicated ALK in neural crest migration and sympathetic neurogenesis. However, very few downstream targets of ALK have been identified. Here, we describe pathological activation of ALK in the neural crest, which promotes proliferation and migration, while preventing differentiation, thus inducing the onset of neuroblastoma. Understanding the effects of ALK activity on neural crest cells will help find new targets for neuroblastoma treatment.
Collapse
|
6
|
Ibrahim Abdul Hakeem AH, Khaled RST, Sherif Ismail M. Expression of Anaplastic Lymphoma Kinase in Astrocytic Tumors (Histopathological and Immunohistochemical Study). Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Astrocytic tumors are the most common primary brain tumors. Glioblastoma is the most common astrocytic tumor representing the highest World Health Organization (WHO) grade (WHO grade IV) with poor prognosis and short survival time. Anaplastic lymphoma kinase (ALK) has a role in embryonic central nervous system development. ALK receptor is thought to contribute to nervous system function, repair, and metabolic homeostasis and is expressed in high-grade tumors like anaplastic large cell lymphoma that makes it a potential target for therapeutic intervention.
AIM: This work aimed to examine the immunohistochemical expression of ALK in astrocytic tumors and its correlation with age, sex, clinical presentation, location, laterality, recurrence, and WHO grade to implicate possible therapeutic potential.
METHODS: This retrospective study was conducted on sixty cases of archived, formalin-fixed, paraffin-embedded tissue blocks that included different subtypes and grades of astrocytic tumors. Immunohistochemistry using ALK monoclonal antibody was performed using a standard avidin-biotin-peroxidase system.
RESULTS: Of the sixty cases, 57 (95%) cases were negative for ALK, while three (5%) cases are positive for ALK; all showed the strong intensity of expression. No statistically significant association was found between ALK expression and astrocytic tumors in addition to other clinical variables of the studied tumors.
CONCLUSIONS: Most cases of astrocytic tumors showed negative ALK expression apart from three positive cases seen in higher WHO grades, especially gliosarcoma. The high number of negative cases for ALK in our study group suggests that ALK expression is not associated with a prognostic significance toward astrocytic tumors whatever its grade.
Collapse
|
7
|
Franceschi E, De Biase D, Di Nunno V, Pession A, Tosoni A, Gatto L, Tallini G, Visani M, Lodi R, Bartolini S, Brandes AA. The clinical and prognostic role of ALK in glioblastoma. Pathol Res Pract 2021; 221:153447. [PMID: 33887544 DOI: 10.1016/j.prp.2021.153447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND anaplastic lymphoma kinase (ALK) overexpression and gene alterations have been detected in several malignancies, with prognostic and therapeutic implications. However, few studies investigated the correlation between ALK altered expression and prognosis in patients with glioblastoma (GBM). METHODS We performed an evaluation of ALK overexpression and structural/quantitative chromosome alterations through immune-histochemical assay (IHC with D5F3 antibody) and fluorescent in situ hybridization (FISH) in patients with isocitrate dehydrogenase (IDH) wild type (wt) GBM. Assuming an ALK overexpression in 20 % of patients we planned a sample of 44 patients to achieve a probability of 90 % to include from 10 % to 30 % of patients with ALK alterations. RESULTS We evaluated 44 patients with IDH wt GBM, treated in our institution and dead due to GBM progression in 2017. ALK overexpression obtained by a composed score (the product of IHC intensity staining and rate of positive cells) was observed in 19 (43 %) patients. FISH analysis showed that 11 patients (25 %) had gene deletion, 2 patients (4.5 %) had monosomy and one patient (2.3 %) presented polysomy. Only one patient (2.3 %) demonstrated ALK rearrangement. There was no statistical difference in median OS between patients with ALK-positive (mOS = 18.9 months) and ALK-negative IHC (mOS = 18.0 months). CONCLUSION We identified some rare previously unreported alterations of ALK gene in patients with IDH wt GBM. In these patients, the ALK overexpression does not influences survival.
Collapse
Affiliation(s)
| | - Dario De Biase
- Department of Pharmacy and Biotechnology (FaBIT) - Molecular Pathology Laboratory, University of Bologna, Bologna, Italy
| | | | - Annalisa Pession
- Department of Pharmacy and Biotechnology (FaBIT) - Molecular Pathology Laboratory, University of Bologna, Bologna, Italy
| | - Alicia Tosoni
- Department of Oncology, AUSL Bologna, Bologna, Italy
| | - Lidia Gatto
- Department of Oncology, AUSL Bologna, Bologna, Italy
| | - Giovanni Tallini
- Molecular Diagnostic Unit, University of Bologna School of Medicine and Surgery, Bologna, Italy
| | - Michela Visani
- Molecular Diagnostic Unit, University of Bologna School of Medicine and Surgery, Bologna, Italy
| | - Raffaele Lodi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Italy
| | | | | |
Collapse
|
8
|
Kawauchi D, Takahashi M, Satomi K, Yamamuro S, Kobayashi T, Uchida E, Honda-Kitahara M, Narita Y, Iwadate Y, Ichimura K, Tomiyama A. The ALK inhibitors, alectinib and ceritinib, induce ALK-independent and STAT3-dependent glioblastoma cell death. Cancer Sci 2021; 112:2442-2453. [PMID: 33728771 PMCID: PMC8177803 DOI: 10.1111/cas.14885] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/11/2021] [Accepted: 03/14/2021] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma (GBM) is the most common, but extremely malignant, brain tumor; thus, the development of novel therapeutic strategies for GBMs is imperative. Many tyrosine kinase inhibitors (TKIs) have been approved for various cancers, yet none has demonstrated clinical benefit against GBM. Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) that is confirmed only during the embryonic development period in humans. In addition, various ALK gene alterations are known to act as powerful oncogenes and therapeutic targets in various tumors. The antitumor activity of various TKIs was tested against three human GBM cell lines (U87MG, LN229, and GSC23), which expressed substantially low ALK levels; second‐generation ALK inhibitors, alectinib and ceritinib, effectively induced GBM cell death. In addition, treatment with either alectinib or ceritinib modulated the activation of various molecules downstream of RTK signaling and induced caspase‐dependent/‐independent cell death mainly by inhibiting signal transducer and activator of transcription 3 activation in human GBM cells. In addition, alectinib and ceritinib also showed antitumor activity against a U87MG cell line with acquired temozolomide resistance. Finally, oral administration of alectinib and ceritinib prolonged the survival of mice harboring intracerebral GBM xenografts compared with controls. These results suggested that treatment with the second‐generation ALK inhibitors, alectinib and ceritinib, might serve as a potent therapeutic strategy against GBM.
Collapse
Affiliation(s)
- Daisuke Kawauchi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masamichi Takahashi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kaishi Satomi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Shun Yamamuro
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurological Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Tatsuya Kobayashi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Eita Uchida
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Hidaka-City, Japan
| | - Mai Honda-Kitahara
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuo Iwadate
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Koichi Ichimura
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Arata Tomiyama
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurosurgery, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
9
|
Das A, Alshareef M, Porto GBF, Infinger LK, Vandergrift WA, Lindhorst SM, Varma AK, Patel SJ, Cachia D. Preconditioning with INC280 and LDK378 drugs sensitizes MGMT-unmethylated glioblastoma to temozolomide: Pre-clinical assessment. J Neurol Sci 2020; 418:117102. [DOI: 10.1016/j.jns.2020.117102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/21/2020] [Accepted: 08/18/2020] [Indexed: 01/29/2023]
|
10
|
Dong Z, Li C, Coates D. PTN-PTPRZ signalling is involved in deer antler stem cell regulation during tissue regeneration. J Cell Physiol 2020; 236:3752-3769. [PMID: 33111346 DOI: 10.1002/jcp.30115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 12/22/2022]
Abstract
A growing deer antler contains a stem cell niche that can drive endochondral bone regeneration at up to 2 cm/day. Pleiotrophin (PTN), as a multifunctional growth factor, is found highly expressed at the messenger RNA level within the active antler stem cell tissues. This study aims to map the expression patterns of PTN protein and its receptors in a growing antler and investigate the effects of PTN on antler stem cells in vitro. Immunohistochemistry was employed to localise PTN/midkine (MDK) and their functional receptors, protein tyrosine phosphatase receptor type Z (PTPRZ), anaplastic lymphoma kinase (ALK), NOTCH2, and integrin αV β3, on serial slides of the antler growth centre. PTN was found to be the dominantly expressed growth factor in the PTN/MDK family. High expression of PTPRZ and ALK co-localised with PTN was found suggesting a potential interaction. The high levels of PTN and PTPRZ reflected the antler stem cell activation status during the regenerative process. When antler stem cells were cultured in vitro under the normoxic condition, no PTN protein was detected and exogenous PTN did not induce differentiation or proliferation but rather stem cell maintenance. Collectively, the antler stem cell niche appears to upregulate PTN and PTPRZ in vivo, and PTN-PTPRZ signalling may be involved in regulating antler stem cell behaviour during rapid antler regeneration.
Collapse
Affiliation(s)
- Zhen Dong
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin, New Zealand
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, China
| | - Dawn Coates
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, Dunedin, New Zealand
| |
Collapse
|
11
|
Yan Y, Wang Y, Liu Y, Chen T, Zhu Y, Li H, Kong F. Long Non-Coding RNA AGAP2-AS1/miR-628-5p/PTN Axis Modulates Proliferation, Migration, Invasion, and Apoptosis of Glioma Cells. Cancer Manag Res 2020; 12:6059-6068. [PMID: 32801858 PMCID: PMC7398883 DOI: 10.2147/cmar.s250890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 06/13/2020] [Indexed: 12/23/2022] Open
Abstract
Purpose Long non-coding RNAs (lncRNAs) have been reported to be involved in a variety of cancers, including glioma. However, the exact role and underlying mechanism of lncRNA AGAP2 antisense RNA 1 (AGAP2-AS1) in glioma have not yet been fully elucidated. Methods The expression levels of AGAP2-AS1, microRNA-628-5p (miR-628-5p) and pleiotrophin (PTN) were measured by quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation, apoptosis, migration and invasion were detected by Cell Counting Kit-8 (CCK-8) assay, flow cytometry, transwell assay, respectively. Western blot assay was used to detect the protein level of PTN. The interaction between miR-628-5p and AGAP2-AS1 or PTN was predicted by bioinformatics software and confirmed by the dual-luciferase reporter and RNA Immunoprecipitation (RIP) assays. Murine xenograft model was established to confirm the role of AGAP2-AS1 in glioma progression in vivo. Results AGAP2-AS1 expression was upregulated in glioma tissues and cells. Knockdown of AGAP2-AS1 inhibited the proliferation, migration and invasion, but facilitated apoptosis in glioma cells. Moreover, AGAP2-AS1 could directly bind to miR-628-5p and its overexpression reversed the anti-tumor effect of miR-628-5p restoration on the progression of glioma cells. In addition, miR-628-5p directly targeted PTN and its inhibition abolished the inhibitory effect of PTN knockdown on the progression of glioma cells. Furthermore, AGAP2-AS1 functioned as a competing endogenous RNA (ceRNA) by sponging miR-628-5p to modulate PTN expression. Besides, AGAP2-AS1 depletion reduced tumor growth by upregulating miR-628-5p and downregulating PTN. Conclusion AGAP2-AS1 knockdown suppressed cell proliferation, migration and invasion but promoted cell apoptosis in glioma cells by regulating miR-628-5p/PTN axis, providing novel avenues for treatment of glioma.
Collapse
Affiliation(s)
- Yang Yan
- Department of Neurosurgery, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China
| | - Yiping Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China
| | - Yuxia Liu
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China
| | - Tao Chen
- Department of Spine Surgery, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China
| | - Yaoli Zhu
- Department of Critical Care Medicine, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China
| | - Huiqing Li
- Department of Neurology, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China
| | - Fangen Kong
- Department of Neurosurgery, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, Guangdong, People's Republic of China
| |
Collapse
|
12
|
Antolčić M, Runje M, Galić N. A simple and sensitive LC-MS/MS method for determination and quantification of potential genotoxic impurities in the ceritinib active pharmaceutical ingredient. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2020; 12:3290-3295. [PMID: 32930193 DOI: 10.1039/d0ay00511h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was used for quantification of four potential genotoxic impurities (PGIs) in the ceritinib active pharmaceutical ingredient. Chromatographic separation was achieved using a YMC-Triart C18 column, with 0.1% formic acid in water as mobile phase A and acetonitrile as mobile phase B in gradient elution mode at a 0.5 mL min-1 flow rate. Quantification of impurities was carried out using triple quadrupole mass detection with electrospray ionization in multiple reaction monitoring mode. The method was fully validated with good linearity over the concentration range of 0.5-5.0 ppm of the ceritinib test concentration for all four PGIs. The correlation coefficient obtained in each case was >0.998. The recoveries were found satisfactory over the range between 83.7 and 107.3% for all selected impurities. The developed method was able to quantitate all four PGIs at a concentration level of 1 ng mL-1 (0.5 ppm with respect to 2 mg mL-1 ceritinib).
Collapse
Affiliation(s)
- Mia Antolčić
- Pliva Croatia, TAPI R&D, Prilaz baruna Filipovića 25, Zagreb 10 000, Croatia
| | - Mislav Runje
- Pliva Croatia, TAPI R&D, Prilaz baruna Filipovića 25, Zagreb 10 000, Croatia
| | - Nives Galić
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, Zagreb 10 000, Croatia.
| |
Collapse
|
13
|
cMyc and ERK activity are associated with resistance to ALK inhibitory treatment in glioblastoma. J Neurooncol 2019; 146:9-23. [DOI: 10.1007/s11060-019-03348-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/15/2019] [Indexed: 12/21/2022]
|
14
|
Zarco N, Norton E, Quiñones-Hinojosa A, Guerrero-Cázares H. Overlapping migratory mechanisms between neural progenitor cells and brain tumor stem cells. Cell Mol Life Sci 2019; 76:3553-3570. [PMID: 31101934 PMCID: PMC6698208 DOI: 10.1007/s00018-019-03149-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/16/2019] [Accepted: 05/13/2019] [Indexed: 01/18/2023]
Abstract
Neural stem cells present in the subventricular zone (SVZ), the largest neurogenic niche of the mammalian brain, are able to self-renew as well as generate neural progenitor cells (NPCs). NPCs are highly migratory and traverse the rostral migratory stream (RMS) to the olfactory bulb, where they terminally differentiate into mature interneurons. NPCs from the SVZ are some of the few cells in the CNS that migrate long distances during adulthood. The migratory process of NPCs is highly regulated by intracellular pathway activation and signaling from the surrounding microenvironment. It involves modulation of cell volume, cytoskeletal rearrangement, and isolation from compact extracellular matrix. In malignant brain tumors including high-grade gliomas, there are cells called brain tumor stem cells (BTSCs) with similar stem cell characteristics to NPCs but with uncontrolled cell proliferation and contribute to tumor initiation capacity, tumor progression, invasion, and tumor maintenance. These BTSCs are resistant to chemotherapy and radiotherapy, and their presence is believed to lead to tumor recurrence at distal sites from the original tumor location, principally due to their high migratory capacity. BTSCs are able to invade the brain parenchyma by utilizing many of the migratory mechanisms used by NPCs. However, they have an increased ability to infiltrate the tight brain parenchyma and utilize brain structures such as myelin tracts and blood vessels as migratory paths. In this article, we summarize recent findings on the mechanisms of cellular migration that overlap between NPCs and BTSCs. A better understanding of the intersection between NPCs and BTSCs will to provide a better comprehension of the BTSCs' invasive capacity and the molecular mechanisms that govern their migration and eventually lead to the development of new therapies to improve the prognosis of patients with malignant gliomas.
Collapse
Affiliation(s)
- Natanael Zarco
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Emily Norton
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, 32224, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, 32224, USA
| | - Alfredo Quiñones-Hinojosa
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Hugo Guerrero-Cázares
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, FL, 32224, USA.
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
15
|
de Nonneville A, Finetti P, Adelaide J, Lambaudie É, Viens P, Gonçalves A, Birnbaum D, Mamessier E, Bertucci F. A Tyrosine Kinase Expression Signature Predicts the Post-Operative Clinical Outcome in Triple Negative Breast Cancers. Cancers (Basel) 2019; 11:E1158. [PMID: 31412533 PMCID: PMC6721506 DOI: 10.3390/cancers11081158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/04/2019] [Accepted: 08/09/2019] [Indexed: 12/13/2022] Open
Abstract
Triple negative breast cancer (TNBC) represent 15% of breast cancers. Histoclinical features and marketed prognostic gene expression signatures (GES) failed to identify good- and poor-prognosis patients. Tyrosine kinases (TK) represent potential prognostic and/or therapeutic targets for TNBC. We sought to define a prognostic TK GES in a large series of TNBC. mRNA expression and histoclinical data of 6379 early BCs were collected from 16 datasets. We searched for a TK-based GES associated with disease-free survival (DFS) and tested its robustness in an independent validation set. A total of 1226 samples were TNBC. In the learning set of samples (N = 825), we identified a 13-TK GES associated with DFS. This GES was associated with cell proliferation and immune response. In multivariate analysis, it outperformed the previously published GESs and classical prognostic factors in the validation set (N = 401), in which the patients classified as "low-risk" had a 73% 5-year DFS versus 53% for "high-risk" patients (p = 1.85 × 10-3). The generation of 100,000 random 13-gene signatures by a resampling scheme showed the non-random nature of our classifier, which was also prognostic for overall survival in multivariate analysis. We identified a robust and non-random 13-TK GES that separated TNBC into subgroups of different prognosis. Clinical and functional validations are warranted.
Collapse
Affiliation(s)
- Alexandre de Nonneville
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Univ, CRCM, CNRS, INSERM, 13000 Marseille, France
| | - Pascal Finetti
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR725, Aix-Marseille Université, 13000 Marseille, France
| | - José Adelaide
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR725, Aix-Marseille Université, 13000 Marseille, France
| | - Éric Lambaudie
- Department of Surgical Oncology, Institut Paoli-Calmettes, Aix-Marseille Univ, CNRS, INSERM, CRCM, 13000 Marseille, France
| | - Patrice Viens
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Univ, CRCM, CNRS, INSERM, 13000 Marseille, France
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Univ, CRCM, CNRS, INSERM, 13000 Marseille, France
| | - Daniel Birnbaum
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR725, Aix-Marseille Université, 13000 Marseille, France
| | - Emilie Mamessier
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR725, Aix-Marseille Université, 13000 Marseille, France
| | - François Bertucci
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille Univ, CRCM, CNRS, INSERM, 13000 Marseille, France.
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR725, Aix-Marseille Université, 13000 Marseille, France.
| |
Collapse
|
16
|
Tanga N, Kuboyama K, Kishimoto A, Kiyonari H, Shiraishi A, Suzuki R, Watanabe T, Fujikawa A, Noda M. The PTN-PTPRZ signal activates the AFAP1L2-dependent PI3K-AKT pathway for oligodendrocyte differentiation: Targeted inactivation of PTPRZ activity in mice. Glia 2019; 67:967-984. [PMID: 30667096 DOI: 10.1002/glia.23583] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022]
Abstract
Protein tyrosine phosphatase receptor type Z (PTPRZ) maintains oligodendrocyte precursor cells (OPCs) in an undifferentiated state. The inhibition of PTPase by its ligand pleiotrophin (PTN) promotes OPC differentiation; however, the substrate molecules of PTPRZ involved in the differentiation have not yet been elucidated in detail. We herein demonstrated that the tyrosine phosphorylation of AFAP1L2, paxillin, ERBB4, GIT1, p190RhoGAP, and NYAP2 was enhanced in OPC-like OL1 cells by a treatment with PTN. AFAP1L2, an adaptor protein involved in the PI3K-AKT pathway, exhibited the strongest response to PTN. PTPRZ dephosphorylated AFAP1L2 at tyrosine residues in vitro and in HEK293T cells. In OL1 cells, the knockdown of AFAP1L2 or application of a PI3K inhibitor suppressed cell differentiation as well as the PTN-induced phosphorylation of AKT and mTOR. We generated a knock-in mouse harboring a catalytically inactive Cys to Ser (CS) mutation in the PTPase domain. The phosphorylation levels of AFAP1L2, AKT, and mTOR were higher, and the expression of oligodendrocyte markers, including myelin basic protein (MBP) and myelin regulatory factor (MYRF), was stronger in CS knock-in brains than in wild-type brains on postnatal day 10; however, these differences mostly disappeared in the adult stage. Adult CS knock-in mice exhibited earlier remyelination after cuprizone-induced demyelination through the accelerated differentiation of OPCs. These phenotypes in CS knock-in mice were similar to those in Ptprz-deficient mice. Therefore, we conclude that the PTN-PTPRZ signal stimulates OPC differentiation partly by enhancing the tyrosine phosphorylation of AFAP1L2 in order to activate the PI3K-AKT pathway.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Carrier Proteins/metabolism
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cuprizone/toxicity
- Cytokines/metabolism
- Demyelinating Diseases/chemically induced
- Demyelinating Diseases/diagnostic imaging
- Disease Models, Animal
- HEK293 Cells
- Humans
- Immunoprecipitation
- In Situ Nick-End Labeling
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microfilament Proteins/metabolism
- Myelin Proteins/metabolism
- Oligodendroglia/physiology
- Proto-Oncogene Proteins c-akt
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics
- Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism
- Signal Detection, Psychological/drug effects
- Signal Detection, Psychological/physiology
- Signal Transduction/physiology
- Transfection
- X-Ray Microtomography
- Red Fluorescent Protein
Collapse
Affiliation(s)
- Naomi Tanga
- Division of Molecular Neurobiology, National Institute for Basic Biology (NIBB), Okazaki, Aichi, Japan
- School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, Japan
| | - Kazuya Kuboyama
- Division of Molecular Neurobiology, National Institute for Basic Biology (NIBB), Okazaki, Aichi, Japan
| | - Ayako Kishimoto
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Nara, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resource Development, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Laboratory for Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Aki Shiraishi
- Laboratory for Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Ryoko Suzuki
- Division of Molecular Neurobiology, National Institute for Basic Biology (NIBB), Okazaki, Aichi, Japan
| | - Toshio Watanabe
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Nara, Japan
| | - Akihiro Fujikawa
- Division of Molecular Neurobiology, National Institute for Basic Biology (NIBB), Okazaki, Aichi, Japan
| | - Masaharu Noda
- Division of Molecular Neurobiology, National Institute for Basic Biology (NIBB), Okazaki, Aichi, Japan
- School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, Japan
- Research Center for Cell Biology, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| |
Collapse
|
17
|
Huang JF, Jiang HY, Cai H, Liu Y, Zhu YQ, Lin SS, Hu TT, Wang TT, Yang WJ, Xiao B, Sun SH, Ma LY, Yin HR, Wang F. Genome-wide screening identifies oncofetal lncRNA Ptn-dt promoting the proliferation of hepatocellular carcinoma cells by regulating the Ptn receptor. Oncogene 2019; 38:3428-3445. [PMID: 30643194 DOI: 10.1038/s41388-018-0643-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 10/04/2018] [Accepted: 11/23/2018] [Indexed: 02/06/2023]
Abstract
Oncofetal genes are genes that express abundantly in both fetal and tumor tissues yet downregulated or undetected in adult tissues, and can be used as tumor markers for cancer diagnosis and treatment. Meanwhile, long noncoding RNAs (lncRNAs) are known to play crucial roles in the pathogenesis of hepatocellular carcinoma (HCC), including tumor growth, proliferation, metastasis, invasion, and recurrence. We performed a genome-wide screening using microarrays to detect the lncRNA expression profiles in fetal livers, adult livers, and liver cancer tissues from mice to identify oncofetal lncRNAs in HCC. From the microarray data analysis, we identified lncRNA Ptn-dt as a possible oncofetal gene. Both in vitro and in vivo experiments results confirmed that overexpression of Ptn-dt significantly promoted the proliferation of mouse HCC cells. RNA pulldown assay showed that Ptn-dt could interact with the HuR protein. Interestingly, miR-96 binds with HuR to maintain its stability as well. Overexpression of lncRNA Ptn-dt led to the downregulation of miR-96, which might be due to the interaction between Ptn-dt and HuR. Meanwhile, previous studies have reported that Ptn can promote tumor growth and vascular abnormalization via anaplastic lymphoma kinase (Alk) signaling. In our study, we found that overexpression of Ptn-dt could promote the expression of Alk through repressing miR-96 via interacting with HuR, thus enhancing the biologic function of Ptn. In summary, a new oncofetal lncRNA Ptn-dt is identified, and it can promote the proliferation of HCC cells by regulating the HuR/miR-96/Alk pathway and Ptn-Alk axis.
Collapse
Affiliation(s)
- Jin-Feng Huang
- Department of Medical Genetics, Second Military Medical University, 200433, Shanghai, China.,Department of Clinical Genetics, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Hong-Yue Jiang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Hui Cai
- Department of General Surgery, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Yan Liu
- Department of Medical Genetics, Second Military Medical University, 200433, Shanghai, China
| | - Yi-Qing Zhu
- Department of Medical Genetics, Second Military Medical University, 200433, Shanghai, China
| | - Sha-Sha Lin
- Center of Reproductive Medicine, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Ting-Ting Hu
- Center of Reproductive Medicine, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Tian-Tian Wang
- Department of Medical Genetics, Second Military Medical University, 200433, Shanghai, China.,Department of Clinical Genetics, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Wen-Jun Yang
- Department of Medical Genetics, Second Military Medical University, 200433, Shanghai, China
| | - Bang Xiao
- Department of Medical Genetics, Second Military Medical University, 200433, Shanghai, China
| | - Shu-Han Sun
- Department of Medical Genetics, Second Military Medical University, 200433, Shanghai, China.,Department of Clinical Genetics, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Li-Ye Ma
- Department of General Surgery, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China.
| | - Hui-Rong Yin
- Center of Reproductive Medicine, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China.
| | - Fang Wang
- Department of Medical Genetics, Second Military Medical University, 200433, Shanghai, China. .,Department of Clinical Genetics, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China.
| |
Collapse
|
18
|
Tang C, Wang M, Wang P, Wang L, Wu Q, Guo W. Neural Stem Cells Behave as a Functional Niche for the Maturation of Newborn Neurons through the Secretion of PTN. Neuron 2018; 101:32-44.e6. [PMID: 30497772 DOI: 10.1016/j.neuron.2018.10.051] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 09/20/2018] [Accepted: 10/29/2018] [Indexed: 10/27/2022]
Abstract
In the neurogenic niches, adult neural stem and/or progenitor cells (NSCs) generate functional neurons throughout life, which has been implicated in learning and memory and affective behaviors. During adult neurogenesis, newborn neurons release feedback signals into the niches to regulate NSC proliferation and differentiation. However, whether and how NSCs contribute to the niche governing newborn neuron development is still unknown. Using a combination of cell ablation, retrovirus-mediated single-cell labeling, and signaling pathway modulation, we show that adult hippocampal NSCs continuously supply pleiotrophin factor to the newborn neurons. Without this feedforward signal, the newborn neurons display defective dendritic development and arborization. Thus, our findings reveal that NSCs behave as a functional niche for newly generated newborn neurons to regulate their maturation.
Collapse
Affiliation(s)
- Changyong Tang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Graduate School, University of the Chinese Academy of Sciences, Beijing 100093, China
| | - Min Wang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Peijian Wang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Graduate School, University of the Chinese Academy of Sciences, Beijing 100093, China
| | - Lei Wang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Graduate School, University of the Chinese Academy of Sciences, Beijing 100093, China
| | - Qingfeng Wu
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Weixiang Guo
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Graduate School, University of the Chinese Academy of Sciences, Beijing 100093, China.
| |
Collapse
|
19
|
Pleiotrophin enhances PDGFB-induced gliomagenesis through increased proliferation of neural progenitor cells. Oncotarget 2018; 7:80382-80390. [PMID: 27806344 PMCID: PMC5348327 DOI: 10.18632/oncotarget.12983] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 10/11/2016] [Indexed: 01/03/2023] Open
Abstract
Pleiotrophin (PTN) augments tumor growth by increasing proliferation of tumor cells and promoting vascular abnormalization, but its role in early gliomagenesis has not been evaluated. Through analysis of publically available datasets, we demonstrate that increased PTN mRNA expression is associated with amplification of chromosome 7, identified as one of the earliest steps in glioblastoma development. To elucidate the role of PTN in tumor initiation we employed the RCAS/tv-a model that allows glioma induction by RCAS-virus mediated expression of oncogenes in neural progenitor cells. Intracranial injection of RCAS-PTN did not induce glioma formation when administrated alone, but significantly enhanced RCAS-platelet derived growth factor (PDGF)B-induced gliomagenesis. PTN co-treatment augmented PDGFB-induced Akt activation in neural progenitor cells in vitro, and enhanced neural sphere size associated with increased proliferation. Our data indicates that PTN expression is associated with chromosome 7 gain, and that PTN enhances PDGFB-induced gliomagenesis by stimulating proliferation of neural progenitor cells.
Collapse
|
20
|
Bao X, Wu J, Sanai N, Li J. A liquid chromatography with tandem mass spectrometry method for quantitating total and unbound ceritinib in patient plasma and brain tumor. J Pharm Anal 2018; 8:20-26. [PMID: 29568664 PMCID: PMC5859147 DOI: 10.1016/j.jpha.2017.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 12/26/2022] Open
Abstract
A rapid, sensitive, and robust reversed-phase liquid chromatography with tandem mass spectrometry method was developed and validated for the determination of total and unbound ceritinib, a second-generation ALK inhibitor, in patient plasma and brain tumor tissue samples. Sample preparation involved simple protein precipitation with acetonitrile. Chromatographic separation was achieved on a Waters ACQUITY UPLC BEH C18 column using a 4-min gradient elution consisting of mobile phase A (0.1% formic acid in water) and mobile phase B (0.1% formic acid in acetonitrile), at a flow rate of 0.4 mL/min. Ceritinib and the internal standard ([13C6]ceritinib) were monitored using multiple reaction monitoring mode under positive electrospray ionization. The lower limit of quantitation (LLOQ) was 1 nM of ceritinib in plasma. The calibration curve was linear over ceritinib concentration range of 1-2000 nM in plasma. The intra- and inter-day precision and accuracy were within the generally accepted criteria for bioanalytical method (<15%). The method was successfully applied to assess ceritinib brain tumor penetration, as assessed by the unbound drug brain concentration to unbound drug plasma concentration ratio, in patients with brain tumors.
Collapse
Affiliation(s)
- Xun Bao
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | - Jianmei Wu
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | - Nader Sanai
- Barrow Neurological Institute, St. Joseph's Hospital&Medical Center, Phoenix, AZ 85013, USA
| | - Jing Li
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
21
|
Junca A, Villalva C, Tachon G, Rivet P, Cortes U, Guilloteau K, Balbous A, Godet J, Wager M, Karayan-Tapon L. Crizotinib targets in glioblastoma stem cells. Cancer Med 2017; 6:2625-2634. [PMID: 28960893 PMCID: PMC5673924 DOI: 10.1002/cam4.1167] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/11/2017] [Accepted: 07/17/2017] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma stem cells (GSCs) are believed to be involved in the mechanisms of tumor resistance, therapeutic failures, and recurrences after conventional glioblastoma therapy. Therefore, elimination of GSCs might be a prerequisite for the development of successful therapeutic strategies. ALK, ROS1, and MET are targeted by Crizotinib, a tyrosine kinase inhibitor which has been approved for treatment of ALK-rearranged non-small-cell lung cancer. In this study we investigated ALK, ROS1, and MET status in nine glioblastoma stem cell lines and tumors from which they arise. Fluorescent in situ hybridization (FISH), Sanger's direct sequencing, and immunohistochemistry were used to screen genomic rearrangements (or amplifications), genomic mutations, and protein expression, respectively. The immunohistochemical and FISH studies revealed no significant dysregulation of ROS1 in GSCs and associated tumors. Neither amplification nor polysomy of ALK was observed in GSC, but weak overexpression was detected by IHC in three of nine GSCs. Similarly, no MET amplification was found by FISH but three GSCs presented significant immunohistochemical staining. No ALK or MET mutation was found by Sanger's direct sequencing. In this study, we show no molecular rearrangement of ALK, ROS1, and MET that would lead us not to propose, as a valid strategy, the use of crizotinib to eradicate GSCs. However, MET was overexpressed in all GSCs with mesenchymal subtype and three GSCs presented an overexpression of ALK. Therefore, our study corroborates the idea that MET and ALK may assume a role in the tumorigenicity of GSC.
Collapse
Affiliation(s)
- Audelaure Junca
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France.,Department of Pathology, University Hospital of Poitiers, Poitiers, F-86021, France.,Medicine and Pharmaceutical Science Faculty, Poitiers University, Poitiers, F-86073, France
| | - Claire Villalva
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France
| | - Gaëlle Tachon
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France.,INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group, University of Poitiers, Poitiers, F-86022, France.,Medicine and Pharmaceutical Science Faculty, Poitiers University, Poitiers, F-86073, France
| | - Pierre Rivet
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France
| | - Ulrich Cortes
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France
| | - Karline Guilloteau
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France
| | - Anaïs Balbous
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France.,INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group, University of Poitiers, Poitiers, F-86022, France.,Medicine and Pharmaceutical Science Faculty, Poitiers University, Poitiers, F-86073, France
| | - Julie Godet
- Department of Pathology, University Hospital of Poitiers, Poitiers, F-86021, France
| | - Michel Wager
- INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group, University of Poitiers, Poitiers, F-86022, France.,Medicine and Pharmaceutical Science Faculty, Poitiers University, Poitiers, F-86073, France.,Department of Neurosurgery, University of Poitiers, Poitiers, F-86021, France
| | - Lucie Karayan-Tapon
- Department of Cancer Biology, University Hospital of Poitiers, Poitiers, F-86021, France.,INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group, University of Poitiers, Poitiers, F-86022, France.,Medicine and Pharmaceutical Science Faculty, Poitiers University, Poitiers, F-86073, France
| |
Collapse
|
22
|
Pleiotrophin promotes chemoresistance to doxorubicin in osteosarcoma by upregulating P-glycoprotein. Oncotarget 2017; 8:63857-63870. [PMID: 28969035 PMCID: PMC5609967 DOI: 10.18632/oncotarget.19148] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 06/10/2017] [Indexed: 12/12/2022] Open
Abstract
Chemoresistance is a major hindrance to successful treatment of osteosarcoma (OS). Pleiotrophin (PTN), a neurotrophic growth factor, has been linked to the malignant characteristics of various cancer types. We retrospectively examined the correlation between PTN expression and chemoresistance in OS in a cohort of 133 OS patients. Immunohistochemistry revealed that PTN expression correlated with the necrosis rate and local OS recurrence. In a prognostic analysis, high PTN expression was associated with poor overall and disease-free survival, and was an independent adverse prognostic factor for disease-free survival. In doxorubicin-treated OS cells, PTN knockdown enhanced cellular chemosensitivity, increased the apoptosis rate and inhibited clone formation, while PTN overexpression had the opposite effects. In a xenograft model, PTN knockdown and overexpression respectively enhanced and reduced cellular sensitivity to doxorubicin. PTN upregulated anaplastic lymphoma kinase (ALK), p-Glycogen Synthase Kinase (GSK)3β, β-catenin and multidrug resistance protein 1/P-glycoprotein (MDR1/P-gp). In rescue assays with the β-catenin inhibitor XAV939 and the MDR1/P-gp inhibitor verapamil, PTN promoted chemoresistance to doxorubicin in OS cells by activating ALK/GSK3β/β-catenin signaling, thereby upregulating MDR1/P-gp. Therefore, PTN could be used as a biomarker predicting chemotherapeutic responses, and downregulating PTN could be a promising therapeutic strategy to prevent chemoresistance in OS patients.
Collapse
|
23
|
Zhang L, Liu X, Liu J, Zhou Z, Song Y, Cao B, An X. miR-182 aids in receptive endometrium development in dairy goats by down-regulating PTN expression. PLoS One 2017; 12:e0179783. [PMID: 28678802 PMCID: PMC5497977 DOI: 10.1371/journal.pone.0179783] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 06/04/2017] [Indexed: 02/04/2023] Open
Abstract
Increasing evidence has shown that miRNAs play important roles in endometrium development during the menstrual cycle in humans and many other animals. Our previous data indicated that miR-182 levels increase 15.55-fold and pleiotrophin (PTN) levels decrease 20.97-fold in the receptive endometrium (RE, D15) compared with the pre-receptive endometrium (PE, D5) in dairy goats. The present study shows that miR-182 is widely expressed in different tissues of dairy goats and that its expression levels are regulated by E2 and P4 in endometrial epithelium cells (EECs). We confirmed that PTN is a target of miR-182 and that miR-182 regulates the protein levels of AKT, Bcl-2, FAS, MAPK, Caspase-3 and SP1 in EECs. Furthermore, miR-182 up-regulates or maintains the expression levels of osteopontin (OPN), cyclooxygenase-2 (COX-2) and prolactin receptor (PRLR) in EECs, suggesting that miR-182 is an important regulatory factor in the construction of endometrial receptivity in dairy goats. In conclusion, miR-182 participates in the development of endometrial receptivity by down-regulating PTN and affecting the expression of select apoptosis-related genes and increasing or maintaining the expression levels of OPN, COX-2 and PRLR in the EECs of dairy goats.
Collapse
Affiliation(s)
- Lei Zhang
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Xiaorui Liu
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Junze Liu
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Zhanqin Zhou
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Binyun Cao
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| |
Collapse
|
24
|
Gugliandolo A, Rajan TS, Scionti D, Diomede F, Bramanti P, Mazzon E, Trubiani O. Reprogramming of Oncogene Expression in Gingival Mesenchymal Stem Cells Following Long-Term Culture In Vitro. Cell Reprogram 2017; 19:159-170. [DOI: 10.1089/cell.2016.0056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
| | | | | | - Francesca Diomede
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University “G. d'Annunzio”, Chieti-Pescara, Chieti, Italy
| | | | | | - Oriana Trubiani
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University “G. d'Annunzio”, Chieti-Pescara, Chieti, Italy
| |
Collapse
|
25
|
Holla VR, Elamin YY, Bailey AM, Johnson AM, Litzenburger BC, Khotskaya YB, Sanchez NS, Zeng J, Shufean MA, Shaw KR, Mendelsohn J, Mills GB, Meric-Bernstam F, Simon GR. ALK: a tyrosine kinase target for cancer therapy. Cold Spring Harb Mol Case Stud 2017; 3:a001115. [PMID: 28050598 PMCID: PMC5171696 DOI: 10.1101/mcs.a001115] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The anaplastic lymphoma kinase (ALK) gene plays an important physiologic role in the development of the brain and can be oncogenically altered in several malignancies, including non-small-cell lung cancer (NSCLC) and anaplastic large cell lymphomas (ALCL). Most prevalent ALK alterations are chromosomal rearrangements resulting in fusion genes, as seen in ALCL and NSCLC. In other tumors, ALK copy-number gains and activating ALK mutations have been described. Dramatic and often prolonged responses are seen in patients with ALK alterations when treated with ALK inhibitors. Three of these—crizotinib, ceritinib, and alectinib—are now FDA approved for the treatment of metastatic NSCLC positive for ALK fusions. However, the emergence of resistance is universal. Newer ALK inhibitors and other targeting strategies are being developed to counteract the newly emergent mechanism(s) of ALK inhibitor resistance. This review outlines the recent developments in our understanding and treatment of tumors with ALK alterations.
Collapse
Affiliation(s)
- Vijaykumar R Holla
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Yasir Y Elamin
- Department of Thoracic/Head and Neck, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ann Marie Bailey
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Amber M Johnson
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Beate C Litzenburger
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Yekaterina B Khotskaya
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Nora S Sanchez
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Jia Zeng
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Md Abu Shufean
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Kenna R Shaw
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - John Mendelsohn
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Gordon B Mills
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Funda Meric-Bernstam
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.,Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - George R Simon
- Department of Thoracic/Head and Neck, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
26
|
Karagkounis G, Stranjalis G, Argyrakos T, Pantelaion V, Mastoris K, Rontogianni D, Komaitis S, Kalamatianos T, Sakas D, Tiniakos D. Anaplastic lymphoma kinase expression and gene alterations in glioblastoma: correlations with clinical outcome. J Clin Pathol 2016; 70:593-599. [DOI: 10.1136/jclinpath-2016-204102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/23/2016] [Accepted: 11/27/2016] [Indexed: 11/04/2022]
Abstract
AimsTo study anaplastic lymphoma kinase (ALK) protein expression and possible underlying gene alterations in glioblastoma (GBM), correlating them with clinical outcome.MethodsWe studied ALK immunohistochemical expression and fluorescent in situ hybridisation (FISH)-detected ALK gene alterations in 51 GBMs (46 isocitrate dehydrogenase-1 (IDH1)R132H-negative and 5 IDH-mutant (IDH1R132H-positive)). We compared two anti-ALK antibodies and immunohistochemical detection systems (5Α4/Nichirei Biosciences, D5F3/Ventana). The results were correlated with tumour cell proliferation and clinical outcome.ResultsIntense granular cytoplasmic ALK immunostaining was observed in 10/51 (19.61%) GBM and correlated with high Ki67 proliferation index; only 1 in 10 ALK-positive cases displayed multiple alk gene signals by FISH. Moderate ALK immunostaining was observed in 21 (41.17%), weak immunostaining in 5 (9.80%) while 15 (29.42%) cases were negative. p53 was expressed in 26/51 GBM (50.9%) (10% cut-off). IDH1R132H-negative GBM showed higher ALK expression compared with IDH-mutant GBM (65.2% vs 20%). ALK overexpression was more common in older patients but did not correlate with other clinicopathological variables or patient overall survival.ConclusionsALK overexpression can be identified in up to 70% of GBMs and does not correlate with underlying alk gene amplification. Despite being more common in rapidly growing, clinically aggressive GBM, ALK overexpression did not show correlation with prognosis in this study.
Collapse
|
27
|
A combination of tyrosine kinase inhibitors, crizotinib and dasatinib for the treatment of glioblastoma multiforme. Oncotarget 2016; 6:37948-64. [PMID: 26517812 PMCID: PMC4741976 DOI: 10.18632/oncotarget.5698] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/06/2015] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor. Despite the advances in surgery, radiotherapy and chemotherapy, patient survival averages only 14.6 months. In most GBM tumors, tyrosine kinases show increased activity and/or expression and actively contribute to the development, recurrence and onset of treatment resistance; making their inhibition an appealing therapeutic strategy. We compared the cytotoxicity of 12 tyrosine kinase inhibitors in vitro. A combination of crizotinib and dasatinib emerged as the most cytotoxic across established and primary human GBM cell lines. The combination treatment induced apoptotic cell death and polyploidy. Furthermore, the combination treatment led to the altered expression and localization of several tyrosine kinase receptors such as Met and EGFR and downstream effectors as such as SRC. Furthermore, the combination treatment reduced the migration and invasion of GBM cells and prevented endothelial cell tube formation in vitro. Overall, our study demonstrated the broad specificity of a combination of crizotinib and dasatinib across multiple GBM cell lines. These findings provide insight into the development of alternative therapy for the treatment of GBM.
Collapse
|
28
|
Chae HS, Kim YM, Chin YW. Atractylodin Inhibits Interleukin-6 by Blocking NPM-ALK Activation and MAPKs in HMC-1. Molecules 2016; 21:molecules21091169. [PMID: 27598116 PMCID: PMC6274166 DOI: 10.3390/molecules21091169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/22/2016] [Accepted: 08/30/2016] [Indexed: 10/26/2022] Open
Abstract
Atractylodin is one of the major constituents of the rhizome of Atractylodes lancea, which is widely used in Korean traditional medicine as a remedy for the treatment of gastritis and gastric ulcers. Despite of a major constituent of widely used botanical to treat inflammatory responses little is known about anti-inflammatory effect of atractylodin in the human mast cell (HMC-1). Hence, we evaluated the effect of atractylodin on the release of IL-6, the involvement of nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) and mitogen-activated protein kinases (MAPKs) in phorbol-12-myristate-13-acetate and A23187-induced HMC-1. In addition, Janus kinase 2 (JAK2), signal transducer and activator of transcription 3 (STAT3), phospholipase C (PLC) gamma 1, and AKT phosphorylation relevant to NPM-ALK signal pathway were assessed. IL-6 levels in the HMC-1 stimulated by phorbol-12-myristate-13-acetate and A23187 were apparently decreased by the treatment of atractylodin. Concurrently, atractylodin not only inhibited the phosphorylation of NPM-ALK, but also suppressed the phosphorylation of JAK2, STAT3, PLC gamma 1, and AKT. Furthermore, the activated mitogen-activated protein kinases (MAPKs) by phorbol-12-myristate-13-acetate and A23187 were inhibited by atractylodin. These results suggested that atractylodin might have a potential regulatory effect on inflammatory mediator expression through blockade of both the phosphorylation of MAPKs and the NPM-ALK signaling pathway.
Collapse
Affiliation(s)
- Hee-Sung Chae
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, 32 Dongguk-lo, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Korea.
| | - Young-Mi Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, 32 Dongguk-lo, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Korea.
| | - Young-Won Chin
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, 32 Dongguk-lo, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Korea.
| |
Collapse
|
29
|
Gonzales CB, De La Chapa JJ, Saikumar P, Singha PK, Dybdal-Hargreaves NF, Chavez J, Horning AM, Parra J, Kirma NB. Co-targeting ALK and EGFR parallel signaling in oral squamous cell carcinoma. Oral Oncol 2016; 59:12-19. [PMID: 27424178 PMCID: PMC5460536 DOI: 10.1016/j.oraloncology.2016.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 04/18/2016] [Accepted: 05/12/2016] [Indexed: 10/21/2022]
Abstract
Squamous cell carcinoma (SCC) comprises 90% of all head and neck cancers and has a poor survival rate due to late-stage disease that is refractive to traditional therapies. Epidermal growth factor receptor (EGFR) is over-expressed in greater than 80% of head and neck SCC (HNSCC). However, EGFR targeted therapies yielded little to no efficacy in clinical trials. This study investigated the efficacy of co-targeting EGFR and the anaplastic lymphoma kinase (ALK) whose promoter is hypomethylated in late-stage oral SCC (OSCC). We observed increased ALK activity in late-stage human OSCC tumors and invasive OSCC cell lines. We also found that while ALK inhibition alone had little effect on proliferation, co-targeting ALK and EGFR significantly reduced OSCC cell proliferation in vitro. Further analysis showed significant efficacy of combined treatment in HSC3-derived xenografts resulting in a 30% decrease in tumor volumes by 14days (p<0.001). Western blot analysis showed that co-targeting ALK and EGFR significantly reduced EGFR phosphorylation (Y1148) in HSC3 cells but not Cal27 cells. ALK and EGFR downstream signaling interactions are also demonstrated by Western blot analysis in which lone EGFR and ALK inhibitors attenuated AKT activity whereas co-targeting ALK and EGFR completely abolished AKT activation. No effects were observed on ERK1/2 activation. STAT3 activity was significantly induced by lone ALK inhibition in HSC3 cells and to a lower extent in Cal27 cells. Together, these data illustrate that ALK inhibitors enhance anti-tumor activity of EGFR inhibitors in susceptible tumors that display increased ALK expression, most likely through abolition of AKT activation.
Collapse
Affiliation(s)
- Cara B Gonzales
- Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX 78229, USA; Comprehensive Dentistry, UTHSCSA Dental School, San Antonio, TX 78229, USA.
| | | | | | | | | | - Jeffery Chavez
- Biochemistry, UTHSCSA Medical School, San Antonio, TX 78229, USA
| | - Aaron M Horning
- Molecular Medicine, UTHSCSA Medical School, San Antonio, TX 78229, USA
| | - Jamie Parra
- Pathology, UTHSCSA Medical School, San Antonio, TX 78229, USA
| | - Nameer B Kirma
- Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX 78229, USA; Molecular Medicine, UTHSCSA Medical School, San Antonio, TX 78229, USA.
| |
Collapse
|
30
|
Aberrant Expression of Anaplastic Lymphoma Kinase in Ovarian Carcinoma Independent of Gene Rearrangement. Int J Gynecol Pathol 2016; 35:337-47. [DOI: 10.1097/pgp.0000000000000260] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Zhang L, Kundu S, Feenstra T, Li X, Jin C, Laaniste L, El Hassan TEA, Ohlin KE, Yu D, Olofsson T, Olsson AK, Pontén F, Magnusson PU, Nilsson KF, Essand M, Smits A, Dieterich LC, Dimberg A. Pleiotrophin promotes vascular abnormalization in gliomas and correlates with poor survival in patients with astrocytomas. Sci Signal 2015; 8:ra125. [PMID: 26645582 DOI: 10.1126/scisignal.aaa1690] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glioblastomas are aggressive astrocytomas characterized by endothelial cell proliferation and abnormal vasculature, which can cause brain edema and increase patient morbidity. We identified the heparin-binding cytokine pleiotrophin as a driver of vascular abnormalization in glioma. Pleiotrophin abundance was greater in high-grade human astrocytomas and correlated with poor survival. Anaplastic lymphoma kinase (ALK), which is a receptor that is activated by pleiotrophin, was present in mural cells associated with abnormal vessels. Orthotopically implanted gliomas formed from GL261 cells that were engineered to produce pleiotrophin showed increased microvessel density and enhanced tumor growth compared with gliomas formed from control GL261 cells. The survival of mice with pleiotrophin-producing gliomas was shorter than that of mice with gliomas that did not produce pleiotrophin. Vessels in pleiotrophin-producing gliomas were poorly perfused and abnormal, a phenotype that was associated with increased deposition of vascular endothelial growth factor (VEGF) in direct proximity to the vasculature. The growth of pleiotrophin-producing GL261 gliomas was inhibited by treatment with the ALK inhibitor crizotinib, the ALK inhibitor ceritinib, or the VEGF receptor inhibitor cediranib, whereas control GL261 tumors did not respond to either inhibitor. Our findings link pleiotrophin abundance in gliomas with survival in humans and mice, and show that pleiotrophin promotes glioma progression through increased VEGF deposition and vascular abnormalization.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Soumi Kundu
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Tjerk Feenstra
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Xiujuan Li
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Chuan Jin
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Liisi Laaniste
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | | | - K Elisabet Ohlin
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Di Yu
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Tommie Olofsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Fredrik Pontén
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Karin Forsberg Nilsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Magnus Essand
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Anja Smits
- Department of Neuroscience, Neurology, Uppsala University, 751 85 Uppsala, Sweden
| | - Lothar C Dieterich
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, The Rudbeck Laboratory, 75185 Uppsala, Sweden.
| |
Collapse
|
32
|
Das A, Cheng RR, Hilbert MLT, Dixon-Moh YN, Decandio M, Vandergrift WA, Banik NL, Lindhorst SM, Cachia D, Varma AK, Patel SJ, Giglio P. Synergistic Effects of Crizotinib and Temozolomide in Experimental FIG-ROS1 Fusion-Positive Glioblastoma. CANCER GROWTH AND METASTASIS 2015; 8:51-60. [PMID: 26648752 PMCID: PMC4667559 DOI: 10.4137/cgm.s32801] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/18/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GB) is the most common malignant brain tumor. Drug resistance frequently develops in these tumors during chemotherapy. Therefore, predicting drug response in these patients remains a major challenge in the clinic. Thus, to improve the clinical outcome, more effective and tolerable combination treatment strategies are needed. Robust experimental evidence has shown that the main reason for failure of treatments is signal redundancy due to coactivation of several functionally linked receptor tyrosine kinases (RTKs), including anaplastic lymphoma kinase (ALK), c-Met (hepatocyte growth factor receptor), and oncogenic c-ros oncogene1 (ROS1: RTK class orphan) fusion kinase FIG (fused in GB)-ROS1. As such, these could be attractive targets for GB therapy. The study subjects consisted of 19 patients who underwent neurosurgical resection of GB tissues. Our in vitro and ex vivo models promisingly demonstrated that treatments with crizotinib (PF-02341066: dual ALK/c-Met inhibitor) and temozolomide in combination induced synergistic antitumor activity on FIG-ROS1-positive GB cells. Our results also showed that ex vivo FIG-ROS1+ slices (obtained from GB patients) when cultured were able to preserve tissue architecture, cell viability, and global gene-expression profiles for up to 14 days. Both in vitro and ex vivo studies indicated that combination blockade of FIG, p-ROS1, p-ALK, and p-Met augmented apoptosis, which mechanistically involves activation of Bim and inhibition of survivin, p-Akt, and Mcl-1 expression. However, it is important to note that we did not see any significant synergistic effect of crizotinib and temozolomide on FIG-ROS1-negative GB cells. Thus, these ex vivo culture results will have a significant impact on patient selection for clinical trials and in predicting response to crizotinib and temozolomide therapy. Further studies in different animal models of FIG-ROS1-positive GB cells are warranted to determine useful therapies for the management of human GBs.
Collapse
Affiliation(s)
- Arabinda Das
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Ron Ron Cheng
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Megan L T Hilbert
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Yaenette N Dixon-Moh
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Michele Decandio
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | | | - Naren L Banik
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA. ; Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Scott M Lindhorst
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - David Cachia
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Abhay K Varma
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Sunil J Patel
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Pierre Giglio
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA. ; Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
33
|
Siraj AK, Beg S, Jehan Z, Prabhakaran S, Ahmed M, R Hussain A, Al-Dayel F, Tulbah A, Ajarim D, Al-Kuraya KS. ALK alteration is a frequent event in aggressive breast cancers. Breast Cancer Res 2015; 17:127. [PMID: 26384210 PMCID: PMC4588266 DOI: 10.1186/s13058-015-0610-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/07/2015] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Breast cancer is the most common female malignancy worldwide and, despite improvements in treatment modalities, there are increased chances of recurrence and metastasis in a substantial number of cases and it remains one of the major causes of mortality among female cancer patients. Anaplastic lymphoma kinase (ALK) gene has been found to be altered in several solid and hematologic tumors. We aimed to comprehensively study the prevalence of ALK expression, and changes in copy number and translocation in a large cohort of breast cancer cases in a Middle Eastern population. METHODS ALK protein expression was investigated by immunohistochemistry and numerical and structural variations of the ALK gene were analyzed by fluorescence in situ hybridization (FISH) in a tissue microarray format in a cohort of more than 1000 Middle Eastern breast cancers. The data were correlated with clinicopathologic parameters and other important molecular biomarkers. RESULTS Immunohistochemical analysis showed ALK overexpression in 36.0 % of the breast cancer patients and gene amplification was present in 13.3 % of cases, seen by FISH analyses. ALK overexpression was significantly associated with ALK gene amplification (p = 0.0031). ALK-overexpressing tumors showed significant association with high-grade tumors (p = 0.0039), ductal histologic subtype (p = 0.0076), triple-negative phenotype (p = 0.0034), and high Ki-67 (p = 0.0001) and p-AKT (p <0.0001). CONCLUSIONS Immunohistochemical analysis showed ALK is overexpressed in a substantial proportion of breast cancers and possibly plays a significant role in the aggressive behavior of this cancer. Gene amplification is hypothesized to be a possible cause for a significant proportion of this overexpression. Based on these findings, a potential role for an ALK inhibitor, as a therapeutic agent targeting aggressive subtypes of breast cancer, merits further investigation.
Collapse
Affiliation(s)
- Abdul K Siraj
- Department of Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Makkah Al Mukarramah Branch Road, Riyadh, 12713, Saudi Arabia.
| | - Shaham Beg
- Department of Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Makkah Al Mukarramah Branch Road, Riyadh, 12713, Saudi Arabia.
| | - Zeenath Jehan
- Department of Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Makkah Al Mukarramah Branch Road, Riyadh, 12713, Saudi Arabia.
| | - Sarita Prabhakaran
- Department of Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Makkah Al Mukarramah Branch Road, Riyadh, 12713, Saudi Arabia.
| | - Maqbool Ahmed
- Department of Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Makkah Al Mukarramah Branch Road, Riyadh, 12713, Saudi Arabia.
| | - Azhar R Hussain
- Department of Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Makkah Al Mukarramah Branch Road, Riyadh, 12713, Saudi Arabia.
| | - Fouad Al-Dayel
- Department of Pathology, King Faisal Specialist Hospital and Research Center, Makkah Al Mukarramah Branch Road, Riyadh, 12713, Saudi Arabia.
| | - Asma Tulbah
- Department of Pathology, King Faisal Specialist Hospital and Research Center, Makkah Al Mukarramah Branch Road, Riyadh, 12713, Saudi Arabia.
| | - Dahish Ajarim
- Oncology Center, King Faisal Specialist Hospital and Research Center, Makkah Al Mukarramah Branch Road, Riyadh, 12713, Saudi Arabia.
| | - Khawla S Al-Kuraya
- Department of Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Makkah Al Mukarramah Branch Road, Riyadh, 12713, Saudi Arabia. .,Department of Pathology, Al-Faisal University, Al Zahrawi Street, Riyadh, 11533, Saudi Arabia.
| |
Collapse
|
34
|
Abstract
Glioblastoma is characterized by microvascular proliferation and a highly abnormal dysfunctional vasculature. The glioblastoma vessels differ significantly from normal brain vessels morphologically, functionally and molecularly. The present review provides a brief overview of the current understanding of the formation, functional abnormalities and specific gene expression of glioblastoma vessels and the consequences of vascular abnormalization for the tumour microenvironment.
Collapse
|
35
|
Mechanisms of Acquired Resistance to ALK Inhibitors and the Rationale for Treating ALK-positive Lung Cancer. Cancers (Basel) 2015; 7:763-83. [PMID: 25941796 PMCID: PMC4491683 DOI: 10.3390/cancers7020763] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 04/14/2015] [Accepted: 04/21/2015] [Indexed: 12/24/2022] Open
Abstract
The discovery of an echinoderm microtubule-associated protein-like 4 (EML4)-anaplastic lymphoma kinase (ALK) fusion gene led to improved clinical outcomes in patients with lung cancer after the development of the first ALK-targeting agent, crizotinib. Some second-generation ALK tyrosine kinase inhibitors (TKIs), which might be more potent than crizotinib or effective on crizotinib-resistant patients, have been developed. Although these ALK-TKIs show an excellent response initially, most patients eventually acquire resistance. Therefore, careful consideration of the resistance mechanisms might lead to superior therapeutic strategies. Here, we summarize the history of ALK-TKIs and their underlying resistance mechanisms in both the preclinical and clinical settings. In addition, we discuss potential future treatment strategies in ALK-TKI-naïve and -resistant patients with lung cancer harboring the EML4-ALK fusion gene.
Collapse
|
36
|
Olsen TK, Panagopoulos I, Meling TR, Micci F, Gorunova L, Thorsen J, Due-Tønnessen B, Scheie D, Lund-Iversen M, Krossnes B, Saxhaug C, Heim S, Brandal P. Fusion genes with ALK as recurrent partner in ependymoma-like gliomas: a new brain tumor entity? Neuro Oncol 2015; 17:1365-73. [PMID: 25795305 DOI: 10.1093/neuonc/nov039] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 02/18/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND We have previously characterized 19 ependymal tumors using Giemsa banding and high-resolution comparative genomic hybridization. The aim of this study was to analyze these tumors searching for fusion genes. METHODS RNA sequencing was performed in 12 samples. Potential fusion transcripts were assessed by seed count and structural chromosomal aberrations. Transcripts of interest were validated using fluorescence in situ hybridization and PCR followed by direct sequencing. RESULTS RNA sequencing identified rearrangements of the anaplastic lymphoma kinase gene (ALK) in 2 samples. Both tumors harbored structural aberrations involving the ALK locus 2p23. Tumor 1 had an unbalanced t(2;14)(p23;q22) translocation which led to the fusion gene KTN1-ALK. Tumor 2 had an interstitial del(2)(p16p23) deletion causing the fusion of CCDC88A and ALK. In both samples, the breakpoint of ALK was located between exons 19 and 20. Both patients were infants and both tumors were supratentorial. The tumors were well demarcated from surrounding tissue and had both ependymal and astrocytic features but were diagnosed and treated as ependymomas. CONCLUSIONS By combining karyotyping and RNA sequencing, we identified the 2 first ever reported ALK rearrangements in CNS tumors. Such rearrangements may represent the hallmark of a new entity of pediatric glioma characterized by both ependymal and astrocytic features. Our findings are of particular importance because crizotinib, a selective ALK inhibitor, has demonstrated effect in patients with lung cancer harboring ALK rearrangements. Thus, ALK emerges as an interesting therapeutic target in patients with ependymal tumors carrying ALK fusions.
Collapse
Affiliation(s)
- Thale Kristin Olsen
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Ioannis Panagopoulos
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Torstein R Meling
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Francesca Micci
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Ludmila Gorunova
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Jim Thorsen
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Bernt Due-Tønnessen
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - David Scheie
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Marius Lund-Iversen
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Bård Krossnes
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Cathrine Saxhaug
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Sverre Heim
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| | - Petter Brandal
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., I.P., F.M., L.G., J.T., S.H., P.B.); Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway (T.K.O., S.H.); Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, Oslo, Norway (T.R.M., B.D.-T.); Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway (M.L.-I., B.K.); Department of Radiology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (C.S.); Department of Pathology, Rigshospitalet, Copenhagen, Denmark (D.S.); Department of Oncology, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway (P.B.)
| |
Collapse
|
37
|
Cazes A, Lopez-Delisle L, Tsarovina K, Pierre-Eugène C, De Preter K, Peuchmaur M, Nicolas A, Provost C, Louis-Brennetot C, Daveau R, Kumps C, Cascone I, Schleiermacher G, Prignon A, Speleman F, Rohrer H, Delattre O, Janoueix-Lerosey I. Activated Alk triggers prolonged neurogenesis and Ret upregulation providing a therapeutic target in ALK-mutated neuroblastoma. Oncotarget 2015; 5:2688-702. [PMID: 24811913 PMCID: PMC4058037 DOI: 10.18632/oncotarget.1883] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Activating mutations of the ALK (Anaplastic lymphoma Kinase) gene have been identified in sporadic and familial cases of neuroblastoma, a cancer of early childhood arising from the sympathetic nervous system (SNS). To decipher ALK function in neuroblastoma predisposition and oncogenesis, we have characterized knock-in (KI) mice bearing the two most frequent mutations observed in neuroblastoma patients. A dramatic enlargement of sympathetic ganglia is observed in AlkF1178L mice from embryonic to adult stages associated with an increased proliferation of sympathetic neuroblasts from E14.5 to birth. In a MYCN transgenic context, the F1178L mutation displays a higher oncogenic potential than the R1279Q mutation as evident from a shorter latency of tumor onset. We show that tumors expressing the R1279Q mutation are sensitive to ALK inhibition upon crizotinib treatment. Furthermore, our data provide evidence that activated ALK triggers RET upregulation in mouse sympathetic ganglia at birth as well as in murine and human neuroblastoma. Using vandetanib, we show that RET inhibition strongly impairs tumor growth in vivo in both MYCN/KI AlkR1279Q and MYCN/KI AlkF1178L mice. Altogether, our findings demonstrate the critical role of activated ALK in SNS development and pathogenesis and identify RET as a therapeutic target in ALK mutated neuroblastoma.
Collapse
Affiliation(s)
- Alex Cazes
- Inserm U830, 26 rue d'Ulm, 75005 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
González-Castillo C, Ortuño-Sahagún D, Guzmán-Brambila C, Pallàs M, Rojas-Mayorquín AE. Pleiotrophin as a central nervous system neuromodulator, evidences from the hippocampus. Front Cell Neurosci 2015; 8:443. [PMID: 25620911 PMCID: PMC4287103 DOI: 10.3389/fncel.2014.00443] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/10/2014] [Indexed: 02/04/2023] Open
Abstract
Pleiotrophin (PTN) is a secreted growth factor, and also a cytokine, associated with the extracellular matrix, which has recently starting to attract attention as a significant neuromodulator with multiple neuronal functions during development. PTN is expressed in several tissues, where its signals are generally related with cell proliferation, growth, and differentiation by acting through different receptors. In Central Nervous System (CNS), PTN exerts post-developmental neurotrophic and -protective effects, and additionally has been involved in neurodegenerative diseases and neural disorders. Studies in Drosophila shed light on some aspects of the different levels of regulatory control of PTN invertebrate homologs. Specifically in hippocampus, recent evidence from PTN Knock-out (KO) mice involves PTN functioning in learning and memory. In this paper, we summarize, discuss, and contrast the most recent advances and results that lead to proposing a PTN as a neuromodulatory molecule in the CNS, particularly in hippocampus.
Collapse
Affiliation(s)
- Celia González-Castillo
- Doctorwado en Ciencias en Biología Molecular en Medicina (DCBMM), CUCS, Universidad de Guadalajara Guadalajara, Jalisco, México
| | - Daniel Ortuño-Sahagún
- Instituto de Investigación en Ciencias Biomédicas (IICB), CUCS, Universidad de Guadalajara, Guadalajara Jalisco, México
| | - Carolina Guzmán-Brambila
- Tecnológico de Monterrey, División de Biotecnología y Salud, Escuela de Medicina, Campus Guadalajara Guadalajara, Jalisco, México
| | - Mercè Pallàs
- Department of Pharmacology and Medical Chemistry, Faculty of Pharmacy School of Pharmacy, Institute of Biomedicine (IBUB), Centros de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), University of Barcelona Barcelona, Spain
| | | |
Collapse
|
39
|
Ceccon M, Mologni L, Giudici G, Piazza R, Pirola A, Fontana D, Gambacorti-Passerini C. Treatment Efficacy and Resistance Mechanisms Using the Second-Generation ALK Inhibitor AP26113 in Human NPM-ALK-Positive Anaplastic Large Cell Lymphoma. Mol Cancer Res 2014; 13:775-83. [PMID: 25421750 DOI: 10.1158/1541-7786.mcr-14-0157] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 11/12/2014] [Indexed: 11/16/2022]
Abstract
UNLABELLED ALK is a tyrosine kinase receptor involved in a broad range of solid and hematologic tumors. Among 70% to 80% of ALK(+) anaplastic large cell lymphomas (ALCL) are caused by the aberrant oncogenic fusion protein NPM-ALK. Crizotinib was the first clinically relevant ALK inhibitor, now approved for the treatment of late-stage and metastatic cases of lung cancer. However, patients frequently develop drug resistance to Crizotinib, mainly due to the appearance of point mutations located in the ALK kinase domain. Fortunately, other inhibitors are available and in clinical trial, suggesting the potential for second-line therapies to overcome Crizotinib resistance. This study focuses on the ongoing phase I/II trial small-molecule tyrosine kinase inhibitor (TKI) AP26113, by Ariad Pharmaceuticals, which targets both ALK and EGFR. Two NPM-ALK(+) human cell lines, KARPAS-299 and SUP-M2, were grown in the presence of increasing concentrations of AP26113, and eight lines were selected that demonstrated resistance. All lines show IC50 values higher (130 to 1,000-fold) than the parental line. Mechanistically, KARPAS-299 populations resistant to AP26113 show NPM-ALK overexpression, whereas SUP-M2-resistant cells harbor several point mutations spanning the entire ALK kinase domain. In particular, amino acid substitutions: L1196M, S1206C, the double F1174V+L1198F and L1122V+L1196M mutations were identified. The knowledge of the possible appearance of new clinically relevant mechanisms of drug resistance is a useful tool for the management of new TKI-resistant cases. IMPLICATIONS This work defines reliable ALCL model systems of AP26113 resistance and provides a valuable tool in the management of all cases of relapse upon NPM-ALK-targeted therapy.
Collapse
Affiliation(s)
- M Ceccon
- Department of Health Science, University of Milano-Bicocca, Monza, Italy.
| | - L Mologni
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - G Giudici
- Tettamanti Research Centre, Pediatric Clinic, University of Milano-Bicocca, Monza, Italy
| | - R Piazza
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - A Pirola
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - D Fontana
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - C Gambacorti-Passerini
- Department of Health Science, University of Milano-Bicocca, Monza, Italy. Section of Haematology, San Gerardo Hospital, Monza, Italy
| |
Collapse
|
40
|
Hugosson F, Sjögren C, Birve A, Hedlund L, Eriksson T, Palmer RH. The Drosophila midkine/pleiotrophin homologues Miple1 and Miple2 affect adult lifespan but are dispensable for alk signaling during embryonic gut formation. PLoS One 2014; 9:e112250. [PMID: 25380037 PMCID: PMC4224452 DOI: 10.1371/journal.pone.0112250] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/10/2014] [Indexed: 01/07/2023] Open
Abstract
Midkine (MDK) and Pleiotrophin (PTN) are small heparin-binding cytokines with closely related structures. The Drosophila genome harbours two genes encoding members of the MDK/PTN family of proteins, known as miple1 and miple2. We have investigated the role of Miple proteins in vivo, in particular with regard to their proposed role as ligands for the Alk receptor tyrosine kinase (RTK). Here we show that Miple proteins are neither required to drive Alk signaling during Drosophila embryogenesis, nor are they essential for development in the fruit fly. Additionally we show that neither MDK nor PTN can activate hALK in vivo when ectopically co-expressed in the fly. In conclusion, our data suggest that Alk is not activated by MDK/PTN related growth factors Miple1 and Miple 2 in vivo.
Collapse
Affiliation(s)
| | - Camilla Sjögren
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Anna Birve
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | | | | - Ruth H. Palmer
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Göteborg, Sweden
- * E-mail:
| |
Collapse
|
41
|
Gorczyński A, Prełowska M, Adam P, Czapiewski P, Biernat W. ALK-positive cancer: still a growing entity. Future Oncol 2014; 10:305-21. [PMID: 24490615 DOI: 10.2217/fon.13.184] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Since the discovery of ALK-positive anaplastic large-cell lymphoma in 1994 many other types of tumors showing ALK expression were disclosed. They form a heterogeneous group, including lung, renal and soft tissue tumors. The biological function of ALK, its role in carcinogenesis and impact exerted on the clinical outcome have been studied by many research groups. New drugs specifically dedicated for ALK inhibition, for example, crizotinib, have been synthesized and have become a viable treatment option for ALK-positive lung adenocarcinoma, and potentially for other ALK-positive cancers. This review summarizes the current state of knowledge concerning ALK-positive neoplasms, focusing on the clinical aspects of the subject.
Collapse
Affiliation(s)
- Adam Gorczyński
- Department of Pathomorphology, Medical University of Gdańsk, Mariana Smoluchowskiego 17, 80-214, Gdańsk, Poland
| | | | | | | | | |
Collapse
|
42
|
Shackelford RE, Vora M, Mayhall K, Cotelingam J. ALK-rearrangements and testing methods in non-small cell lung cancer: a review. Genes Cancer 2014; 5:1-14. [PMID: 24955213 PMCID: PMC4063252 DOI: 10.18632/genesandcancer.3] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 04/22/2014] [Indexed: 01/25/2023] Open
Abstract
The anaplastic lymphoma tyrosine kinase (ALK) gene was first described as a driver mutation in anaplastic non-Hodgkin's lymphoma. Dysregulated ALK expression is now an identified driver mutation in nearly twenty different human malignancies, including 4-9% of non-small cell lung cancers (NSCLC). The tyrosine kinase inhibitor crizotinib is more effective than standard chemotherapeutic agents in treating ALK positive NSCLC, making molecular diagnostic testing for dysregulated ALK expression a necessary step in identifying optimal treatment modalities. Here we review ALKmediated signal transduction pathways and compare the molecular protocols used to identify dysregulated ALK expression in NSCLC. We also discuss the use of crizotinib and second generation ALK tyrosine kinase inhibitors in the treatment of ALK positive NSCLC, and the known mechanisms of crizotinib resistance in NSCLC.
Collapse
Affiliation(s)
| | - Moiz Vora
- LSU Health Shreveport, Department of Pathology, Shreveport, LA, USA
| | - Kim Mayhall
- Tulane University School of Medicine, New Orleans, LA, USA
| | - James Cotelingam
- LSU Health Shreveport, Department of Pathology, Shreveport, LA, USA
| |
Collapse
|
43
|
Gombash SE, Manfredsson FP, Mandel RJ, Collier TJ, Fischer DL, Kemp CJ, Kuhn NM, Wohlgenant SL, Fleming SM, Sortwell CE. Neuroprotective potential of pleiotrophin overexpression in the striatonigral pathway compared with overexpression in both the striatonigral and nigrostriatal pathways. Gene Ther 2014; 21:682-93. [PMID: 24807806 DOI: 10.1038/gt.2014.42] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 02/23/2014] [Accepted: 03/26/2014] [Indexed: 01/06/2023]
Abstract
Intrastriatal injection of recombinant adeno-associated viral vector serotype 2/1 (rAAV2/1) to overexpress the neurotrophic factor pleiotrophin (PTN) provides neuroprotection for tyrosine hydroxylase immunoreactive (THir) neurons in the substantia nigra pars compacta (SNpc), increases THir neurite density in the striatum (ST) and reverses functional deficits in forepaw use following 6-hydroxydopamine (6-OHDA) toxic insult. Glial cell line-derived neurotrophic factor (GDNF) gene transfer studies suggest that optimal neuroprotection is dependent on the site of nigrostriatal overexpression. The present study was conducted to determine whether enhanced neuroprotection could be accomplished via simultaneous rAAV2/1 PTN injections into the ST and SN compared with ST injections alone. Rats were unilaterally injected in the ST alone or injected in both the ST and SN with rAAV2/1 expressing either PTN or control vector. Four weeks later, all rats received intrastriatal injections of 6-OHDA. Rats were euthanized 6 or 16 weeks relative to 6-OHDA injection. A novel selective total enumeration method to estimate nigral THir neuron survival was validated to maintain the accuracy of stereological assessment. Long-term nigrostriatal neuroprotection and functional benefits were only observed in rats in which rAAV2/1 PTN was injected into the ST alone. Results suggest that superior preservation of the nigrostriatal system is provided by PTN overexpression delivered to the ST and restricted to the ST and SN pars reticulata and is not improved with overexpression of PTN within SNpc neurons.
Collapse
Affiliation(s)
- S E Gombash
- 1] Graduate Program in Neuroscience, University of Cincinnati, Cincinnati, OH, USA [2] Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - F P Manfredsson
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - R J Mandel
- Department of Neuroscience, Powell Gene Therapy Center, McKnight Brain Institute, University of Florida College of Medicine, Gainesville, FL, USA
| | - T J Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - D L Fischer
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - C J Kemp
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - N M Kuhn
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - S L Wohlgenant
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - S M Fleming
- Departments of Psychology and Neurology, University of Cincinnati, Cincinnati, OH, USA
| | - C E Sortwell
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
44
|
Fan JB, Liu W, Yuan K, Zhu XH, Xu DW, Chen JJ, Cui ZM. EGFR trans-activation mediates pleiotrophin-induced activation of Akt and Erk in cultured osteoblasts. Biochem Biophys Res Commun 2014; 447:425-30. [PMID: 24727451 DOI: 10.1016/j.bbrc.2014.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 04/02/2014] [Indexed: 12/28/2022]
Abstract
Pleiotrophin (Ptn) plays an important role in bone growth through regulating osteoblasts' functions. The underlying signaling mechanisms are not fully understood. In the current study, we found that Ptn induced heparin-binding epidermal growth factor (HB-EGF) release to trans-activate EGF-receptor (EGFR) in both primary osteoblasts and osteoblast-like MC3T3-E1 cells. Meanwhile, Ptn activated Akt and Erk signalings in cultured osteoblasts. The EGFR inhibitor AG1478 as well as the monoclonal antibody against HB-EGF (anti-HB-EGF) significantly inhibited Ptn-induced EGFR activation and Akt and Erk phosphorylations in MC3T3-E1 cells and primary osteoblasts. Further, EGFR siRNA depletion or dominant negative mutation suppressed also Akt and Erk activation in MC3T3-E1 cells. Finally, we observed that Ptn increased alkaline phosphatase (ALP) activity and inhibited dexamethasone (Dex)-induced cell death in both MC3T3-E1 cells and primary osteoblasts, such effects were alleviated by AG1478 or anti-HB-EGF. Together, these results suggest that Ptn-induced Akt/Erk activation and some of its pleiotropic functions are mediated by EGFR trans-activation in cultured osteoblasts.
Collapse
Affiliation(s)
- Jian-Bo Fan
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, North Hai-er-xiang Road 6, Nantong 226001, Jiangsu, People's Republic of China
| | - Wei Liu
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, North Hai-er-xiang Road 6, Nantong 226001, Jiangsu, People's Republic of China
| | - Kun Yuan
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, North Hai-er-xiang Road 6, Nantong 226001, Jiangsu, People's Republic of China
| | - Xin-Hui Zhu
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, North Hai-er-xiang Road 6, Nantong 226001, Jiangsu, People's Republic of China
| | - Da-Wei Xu
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, North Hai-er-xiang Road 6, Nantong 226001, Jiangsu, People's Republic of China
| | - Jia-Jia Chen
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, North Hai-er-xiang Road 6, Nantong 226001, Jiangsu, People's Republic of China
| | - Zhi-Ming Cui
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, North Hai-er-xiang Road 6, Nantong 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
45
|
Murga-Zamalloa C, Lim MS. ALK-driven tumors and targeted therapy: focus on crizotinib. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2014; 7:87-94. [PMID: 24715763 PMCID: PMC3977456 DOI: 10.2147/pgpm.s37504] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Receptor tyrosine kinases have emerged as promising therapeutic targets for a diverse set of tumors. Overactivation of the tyrosine kinase anaplastic lymphoma kinase (ALK) has been reported in several types of malignancies such as anaplastic large cell lymphoma, inflammatory myofibroblastic tumor, neuroblastoma, and non-small-cell lung carcinoma. Further characterization of the molecular role of ALK has revealed an oncogenic signaling signature that results in tumor dependence on ALK. ALK-positive tumors display a different behavior than their ALK-negative counterparts; however, the specific role of ALK in some of these tumors remains to be elucidated. Although more studies are required to establish selective targeting of ALK as a definitive therapeutic option, initial trials have shown extraordinary results in the majority of cases.
Collapse
Affiliation(s)
| | - Megan S Lim
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
46
|
ALK: Anaplastic lymphoma kinase. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
47
|
ALK as a paradigm of oncogenic promiscuity: different mechanisms of activation and different fusion partners drive tumors of different lineages. Cancer Genet 2013; 206:357-73. [PMID: 24091028 DOI: 10.1016/j.cancergen.2013.07.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/20/2013] [Accepted: 07/22/2013] [Indexed: 12/23/2022]
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase protein implicated in a variety of hematological malignancies and solid tumors. Since the identification of the ALK gene in 1994 as the target of the t(2;5) chromosomal translocation in anaplastic large cell lymphoma, ALK has been proven a remarkably promiscuous oncogene. ALK contributes to the development of a notable assortment of tumor types from different lineages, including hematolymphoid, mesenchymal, epithelial and neural tumors, through a variety of genetic mechanisms: gene fusions, activating point mutations, and gene amplification. Recent developments led to significant diagnostic and therapeutic advances, including efficient diagnostic tests and ALK-targeting agents readily available in the clinical setting. This review addresses some therapeutic considerations of ALK-targeted agents and the biologic implications of ALK oncogenic promiscuity, but the main points discussed are: 1) the variety of mechanisms that result in activation of the ALK oncogene, with emphasis on the promiscuous partnerships demonstrated in chromosomal rearrangements; 2) the diversity of tumor types of different lineages in which ALK has been implicated as a pathogenic driver; and 3) the different diagnostic tests available to identify ALK-driven tumors, and their respective indications.
Collapse
|
48
|
Deuel TF. Anaplastic lymphoma kinase: "Ligand Independent Activation" mediated by the PTN/RPTPβ/ζ signaling pathway. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2219-23. [PMID: 23777859 DOI: 10.1016/j.bbapap.2013.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 06/06/2013] [Accepted: 06/06/2013] [Indexed: 01/26/2023]
Abstract
Anaplastic lymphoma kinase is essential in early development, differentiation, and maintenance of cell survival; nevertheless, the mechanism to activate ALK has remained elusive. ALK has remained an "Orphan Receptor." The studies cited below describe a unique mechanism termed "Ligand Independent Activation." It is shown that activation of ALK results when the cytokine pleiotrophin (PTN) interacts with its receptor, the receptor protein tyrosine phosphatase β/ζ (RPTPβ/ζ). Pleiotrophin inactivates the catalytic activity of RPTPβ/ζ, which, when not inactivated, dephosphorylates phosphotyrosine sites in the activation domain of ALK; as a consequence of the inactivation of RPTPβ/ζ by PTN, autophosphorylation and autoactivation of ALK rapidly follow. The PTN/RPTPβ/ζ signaling pathway thus regulates the catalytic activity of ALK and tyrosine phosphorylation levels of ALK downstream target proteins. Furthermore, since ALK is only one of the key ALK phosphoproteins targeted by the PTN/RPTPβ/ζ signaling pathway, the PTN/RPTPβ/ζ signaling pathway has the potential to coordinately regulate tyrosine phosphorylation of other different key proteins in multiple cellular compartments. This article is part of a Special Issue entitled: Emerging recognition and activation mechanisms of receptor tyrosine kinases.
Collapse
Affiliation(s)
- Thomas F Deuel
- The Scripps Research Institute, La Jolla, CA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
The pleiotrophin-ALK axis is required for tumorigenicity of glioblastoma stem cells. Oncogene 2013; 33:2236-44. [PMID: 23686309 DOI: 10.1038/onc.2013.168] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 03/23/2013] [Accepted: 03/25/2013] [Indexed: 12/18/2022]
Abstract
Increasing evidence suggests that brain tumors arise from the transformation of neural stem/precursor/progenitor cells. Much current research on human brain tumors is focused on the stem-like properties of glioblastoma. Here we show that anaplastic lymphoma kinase (ALK) and its ligand pleiotrophin are required for the self-renewal and tumorigenicity of glioblastoma stem cells (GSCs). Furthermore, we demonstrate that pleiotrophin is transactivated directly by SOX2, a transcription factor essential for the maintenance of both neural stem cells and GSCs. We speculate that the pleiotrophin-ALK axis may be a promising target for the therapy of glioblastoma.
Collapse
|
50
|
Expression of anaplastic lymphoma kinase in Merkel cell carcinomas. Hum Pathol 2013; 44:1656-64. [PMID: 23574788 DOI: 10.1016/j.humpath.2012.11.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 11/13/2012] [Accepted: 11/13/2012] [Indexed: 11/23/2022]
Abstract
This study examines the presence of anaplastic lymphoma kinase protein and anaplastic lymphoma kinase gene rearrangements in Merkel cell carcinomas. A total of 32 cases of Merkel cell carcinomas and 12 cases of small cell lung carcinomas were analyzed. Immunohistochemistry was performed using 3 different anaplastic lymphoma kinase antibody clones (D5F3, 5A4, and anaplastic lymphoma kinase 1). Tumors were divided into high (intensity score 2-3+ in ≥25% of the tumor cells) and low expressors (all other positive expression patterns). Anaplastic lymphoma kinase reactivity in Merkel cell carcinoma was observed in 93.8% (30/32) with clone D5F3, 87.5% (28/32) with clone 5A4, and 12.5% (4/32) with clone anaplastic lymphoma kinase 1. One small cell lung carcinoma (1/12; 8.3%) showed anaplastic lymphoma kinase low expression with clone D5F3. Anaplastic lymphoma kinase high expression was observed in 81.3% (26/32) of the Merkel cell carcinomas with clone D5F3, 71.9% (23/32) with clone 5A4, and none with clone anaplastic lymphoma kinase 1. The specificity of anaplastic lymphoma kinase expression in Merkel cell carcinoma versus small cell lung carcinoma was 91.7% with clone D5F3 and 100% with the clones 5A4 and anaplastic lymphoma kinase 1. Interphase fluorescence in situ hybridization with the anaplastic lymphoma kinase dual-color, break-apart rearrangement probe was performed on 10 randomly selected Merkel cell carcinoma anaplastic lymphoma kinase high expressors. No rearrangement or other cytogenetic aberration of the anaplastic lymphoma kinase gene locus was identified. In conclusion, the anaplastic lymphoma kinase protein was detected with high frequency in Merkel cell carcinomas and was useful in distinguishing Merkel cell carcinoma from small cell lung carcinoma. No correlation with anaplastic lymphoma kinase rearrangement was found. Our findings could have important therapeutic consequences for patients, but the role of anaplastic lymphoma kinase in the pathogenesis of Merkel cell carcinoma needs to be further elucidated.
Collapse
|