851
|
Gurunathan S, Kim JH. Graphene Oxide-Silver Nanoparticles Nanocomposite Stimulates Differentiation in Human Neuroblastoma Cancer Cells (SH-SY5Y). Int J Mol Sci 2017; 18:E2549. [PMID: 29182571 PMCID: PMC5751152 DOI: 10.3390/ijms18122549] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/17/2017] [Accepted: 11/23/2017] [Indexed: 12/16/2022] Open
Abstract
Recently, graphene and graphene related nanocomposite receive much attention due to high surface-to-volume ratio, and unique physiochemical and biological properties. The combination of metallic nanoparticles with graphene-based materials offers a promising method to fabricate novel graphene-silver hybrid nanomaterials with unique functions in biomedical nanotechnology, and nanomedicine. Therefore, this study was designed to prepare graphene oxide (GO) silver nanoparticles (AgNPs) nanocomposite (GO-AgNPs) containing two different nanomaterials in single platform with distinctive properties using luciferin as reducing agents. In addition, we investigated the effect of GO-AgNPs on differentiation in SH-SY5Y cells. The synthesized GO-AgNPs were characterized by ultraviolet-visible absorption spectroscopy (UV-vis), X-ray diffraction (XRD), scanning electron microscopy (SEM), transmission electron microscopy (TEM) and Raman spectroscopy. The differentiation was confirmed by series of cellular and biochemical assays. The AgNPs were distributed uniformly on the surface of graphene oxide with an average size of 25 nm. As prepared GO-AgNPOs induces differentiation by increasing the expression of neuronal differentiation markers and decreasing the expression of stem cell markers. The results indicated that the redox biology involved the expression of various signaling molecules, which play an important role in differentiation. This study suggests that GO-AgNP nanocomposite could stimulate differentiation of SH-SY5Y cells. Furthermore, understanding the mechanisms of differentiation of neuroblastoma cells could provide new strategies for cancer and stem cell therapies. Therefore, these studies suggest that GO-AgNPs could target specific chemotherapy-resistant cells within a tumor.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
852
|
Umapathy G, Guan J, Gustafsson DE, Javanmardi N, Cervantes-Madrid D, Djos A, Martinsson T, Palmer RH, Hallberg B. MEK inhibitor trametinib does not prevent the growth of anaplastic lymphoma kinase (ALK)-addicted neuroblastomas. Sci Signal 2017; 10:10/507/eaam7550. [PMID: 29184034 DOI: 10.1126/scisignal.aam7550] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Activation of the RAS-RAF-MEK-ERK signaling pathway is implicated in driving the initiation and progression of multiple cancers. Several inhibitors targeting the RAS-MAPK pathway are clinically approved as single- or polyagent therapies for patients with specific types of cancer. One example is the MEK inhibitor trametinib, which is included as a rational polytherapy strategy for treating EML4-ALK-positive, EGFR-activated, or KRAS-mutant lung cancers and neuroblastomas that also contain activating mutations in the RAS-MAPK pathway. In addition, in neuroblastoma, a heterogeneous disease, relapse cases display an increased rate of mutations in ALK, NRAS, and NF1, leading to increased activation of RAS-MAPK signaling. Co-targeting ALK and the RAS-MAPK pathway is an attractive option, because monotherapies have not yet produced effective results in ALK-addicted neuroblastoma patients. We evaluated the response of neuroblastoma cell lines to MEK-ERK pathway inhibition by trametinib. In contrast to RAS-MAPK pathway-mutated neuroblastoma cell lines, ALK-addicted neuroblastoma cells treated with trametinib showed increased activation (inferred by phosphorylation) of the kinases AKT and ERK5. This feedback response was mediated by the mammalian target of rapamycin complex 2-associated protein SIN1, resulting in increased survival and proliferation that depended on AKT signaling. In xenografts in mice, trametinib inhibited the growth of EML4-ALK-positive non-small cell lung cancer and RAS-mutant neuroblastoma but not ALK-addicted neuroblastoma. Thus, our results advise against the seemingly rational option of using MEK inhibitors to treat ALK-addicted neuroblastoma.
Collapse
Affiliation(s)
- Ganesh Umapathy
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden
| | - Jikui Guan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden
| | - Dan E Gustafsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden
| | - Niloufar Javanmardi
- Department of Clinical Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden
| | - Diana Cervantes-Madrid
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden
| | - Anna Djos
- Department of Clinical Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden
| | - Tommy Martinsson
- Department of Clinical Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden
| | - Ruth H Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden.
| |
Collapse
|
853
|
Liu SL, Liu Z, Zhang LD, Zhu HQ, Guo JH, Zhao M, Wu YL, Liu F, Gao FH. GSK3β-dependent cyclin D1 and cyclin E1 degradation is indispensable for NVP-BEZ235 induced G0/G1 arrest in neuroblastoma cells. Cell Cycle 2017; 16:2386-2395. [PMID: 28980866 DOI: 10.1080/15384101.2017.1383577] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cyclin D1 and cyclin E1, as vital regulatory factors of G1-S phase cell cycle progression, are frequently constitutive expressed and associated with pathogenesis and tumorigenesis in most human cancers and they have been regarded as promising targets for cancer therapy. In this study, we established NVP-BEZ235, a potent dual kinase inhibitor, could induce neuroblastoma cells proliferation inhibition without apoptosis activation. Moreover, we showed NVP-BEZ235 could induce neuroblastoma cells arrested at G0/G1 phase accompanied with significant reduction of the cyclin D1 and E1 proteins in a dose dependent manner at nanomole concentration. Additionally we found that GSK3β was dephosphorylated and activated by NVP-BEZ235 and then triggered cyclin D1 and cyclin E1 degradation through ubiquitination proteasome pathway, based on the evidences that NVP-BEZ235 induced downregulation of cyclin D1 and cyclin E1 were obviously recovered by proteasome inhibitor and the blockade of GSK3β contributed to remarkable rescue of cyclin D1 and cyclin E1. Analogous results about its anti-proliferation effects and molecular mechanism were observed on neuroblastoma xenograft mouse model in vivo. Therefore, these results indicate that NVP-BEZ235-induced cyclin D1 and cyclin E1 degradation, which happened through activating GSK3β, and GSK3β-dependent down-regulation of cyclin D1 and cyclin E1 should be available for anticancer therapeutics.
Collapse
Affiliation(s)
- Shan-Ling Liu
- a Department of Oncology , Shanghai 9th People's Hospital , Shanghai Jiao Tong University School of Medicine , 639 Zhi Zao Ju Rd, Shanghai , China
| | - Zhen Liu
- a Department of Oncology , Shanghai 9th People's Hospital , Shanghai Jiao Tong University School of Medicine , 639 Zhi Zao Ju Rd, Shanghai , China.,b Department of Clinical Laboratory , Shanghai Pudong Hospital , Fudan University Pudong Medical Center , 2800 Gongwei Road, Pudong, Shanghai , China
| | - Li-Di Zhang
- a Department of Oncology , Shanghai 9th People's Hospital , Shanghai Jiao Tong University School of Medicine , 639 Zhi Zao Ju Rd, Shanghai , China
| | - Han-Qing Zhu
- a Department of Oncology , Shanghai 9th People's Hospital , Shanghai Jiao Tong University School of Medicine , 639 Zhi Zao Ju Rd, Shanghai , China
| | - Jia-Hui Guo
- a Department of Oncology , Shanghai 9th People's Hospital , Shanghai Jiao Tong University School of Medicine , 639 Zhi Zao Ju Rd, Shanghai , China
| | - Mei Zhao
- c Department of Reproductive Medicine , Shanghai First Maternity and Infant Hospital , Tongji University School of Medicine , Shanghai , China
| | - Ying-Li Wu
- d Dept. of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education , Shanghai Jiao-Tong University School of Medicine (SJTU-SM) , Shanghai , China
| | - Feng Liu
- a Department of Oncology , Shanghai 9th People's Hospital , Shanghai Jiao Tong University School of Medicine , 639 Zhi Zao Ju Rd, Shanghai , China
| | - Feng-Hou Gao
- a Department of Oncology , Shanghai 9th People's Hospital , Shanghai Jiao Tong University School of Medicine , 639 Zhi Zao Ju Rd, Shanghai , China
| |
Collapse
|
854
|
Claeys S, Denecker G, Cannoodt R, Kumps C, Durinck K, Speleman F, De Preter K. Early and late effects of pharmacological ALK inhibition on the neuroblastoma transcriptome. Oncotarget 2017; 8:106820-106832. [PMID: 29290991 PMCID: PMC5739776 DOI: 10.18632/oncotarget.22423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/17/2017] [Indexed: 11/25/2022] Open
Abstract
Background Neuroblastoma is an aggressive childhood malignancy of the sympathetic nervous system. Despite multi-modal therapy, survival of high-risk patients remains disappointingly low, underscoring the need for novel treatment strategies. The discovery of ALK activating mutations opened the way to precision treatment in a subset of these patients. Previously, we investigated the transcriptional effects of pharmacological ALK inhibition on neuroblastoma cell lines, six hours after TAE684 administration, resulting in the 77-gene ALK signature, which was shown to gradually decrease from 120 minutes after TAE684 treatment, to gain deeper insight into the molecular effects of oncogenic ALK signaling. Aim Here, we further dissected the transcriptional dynamic profiles of neuroblastoma cells upon TAE684 treatment in a detailed timeframe of ten minutes up to six hours after inhibition, in order to identify additional early targets for combination treatment. Results We observed an unexpected initial upregulation of positively regulated MYCN target genes following subsequent downregulation of overall MYCN activity. In addition, we identified adrenomedullin (ADM), previously shown to be implicated in sunitinib resistance, as the earliest response gene upon ALK inhibition. Conclusions We describe the early and late effects of ALK inhibitor TAE684 treatment on the neuroblastoma transcriptome. The observed unexpected upregulation of ADM warrants further investigation in relation to putative ALK resistance in neuroblastoma patients currently undergoing ALK inhibitor treatment.
Collapse
Affiliation(s)
- Shana Claeys
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Geertrui Denecker
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Robrecht Cannoodt
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Bioinformatics Institute Ghent From Nucleotides to Networks, Ghent, Belgium.,Data Mining and Modelling for Biomedicine group, VIB Inflammation Research Center, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University, Ghent, Belgium
| | - Candy Kumps
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Department of Uro-Gynaecology, Ghent University Hospital, Ghent, Belgium
| | - Kaat Durinck
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Frank Speleman
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Katleen De Preter
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| |
Collapse
|
855
|
Ripperger T, Wimmer K, Kratz C. Seltene Tumordispositionssyndrome mit Manifestation im Kindesalter. MED GENET-BERLIN 2017. [DOI: 10.1007/s11825-017-0150-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Zusammenfassung
Bei etwa 7–10 % der pädiatrischen Krebspatienten werden zugrunde liegende Tumordispositionssyndrome (TDS) vermutet. Das Erkennen von TDS hat klinische Implikationen für die Krebsprävention und -früherkennung, die Krebstherapie und -nachsorge, die psychosoziale Unterstützung sowie die Beratung von Angehörigen und Identifizierung weiterer Anlageträger in den betroffenen Familien. Hinweise auf das Vorliegen eines TDS anhand von Eigen- und Familienanamnese, Untersuchungsbefund sowie gegebenenfalls Tumorhistologie und -genetik müssen daher möglichst früh erkannt werden, um bei Verdacht auf Vorliegen eines TDS eine humangenetische Beratung und gegebenenfalls genetische Diagnostik zu veranlassen. Wissenschaftliche Untersuchungen zu TDS liefern Einblicke in die Biologie der Gewebe- und Tumorentwicklung und weisen auf mögliche Ansatzpunkte zielgerichteter Therapien hin. Die vorliegende Arbeit gibt eine Übersicht über TDS mit erhöhtem Risiko für Wilms-Tumoren (Nephroblastome), Neuroblastome oder Medulloblastome. Zusätzlich werden zwei vergleichsweise neu beschriebene Syndrome mit breitem Neoplasiespektrum erläutert: die konstitutionelle Mismatch-Reparatur-Defizienz (CMMRD) und das DICER1-Syndrom. Neben der Erläuterung der klinischen Charakteristika und der genetischen Grundlagen werden für die tägliche Praxis Hinweise zur Indikation von genetischen Untersuchungen und Früherkennung bei TDS aufgeführt. Die Betreuung der Betroffenen und ihrer Angehörigen sollte möglichst interdisziplinär erfolgen. Forschung zu TDS, zum Beispiel im Rahmen von Registern für TDS, ist essenziell, um langfristig die medizinische Versorgung von Menschen zu verbessern, die bedingt durch konstitutionelle genetische Veränderungen ein erhöhtes Krebsrisiko haben.
Collapse
Affiliation(s)
- Tim Ripperger
- Aff1 0000 0000 9529 9877 grid.10423.34 Institut für Humangenetik Medizinische Hochschule Hannover Hannover Deutschland
| | - Katharina Wimmer
- Aff2 0000 0000 8853 2677 grid.5361.1 Department für Medizinische Genetik, Molekulare und Klinische Pharmakologie Medizinische Universität Innsbruck Innsbruck Österreich
| | - Christian Kratz
- Aff3 0000 0000 9529 9877 grid.10423.34 Klinik für Pädiatrische Hämatologie und Onkologie Medizinische Hochschule Hannover Carl-Neuberg-Str. 1 30625 Hannover Deutschland
| |
Collapse
|
856
|
Novel Mechanisms of ALK Activation Revealed by Analysis of the Y1278S Neuroblastoma Mutation. Cancers (Basel) 2017; 9:cancers9110149. [PMID: 29084134 PMCID: PMC5704167 DOI: 10.3390/cancers9110149] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/09/2017] [Accepted: 10/23/2017] [Indexed: 12/20/2022] Open
Abstract
Numerous mutations have been observed in the Anaplastic Lymphoma Kinase (ALK) receptor tyrosine kinase (RTK) in both germline and sporadic neuroblastoma. Here, we have investigated the Y1278S mutation, observed in four patient cases, and its potential importance in the activation of the full length ALK receptor. Y1278S is located in the 1278-YRASYY-1283 motif of the ALK activation loop, which has previously been reported to be important in the activation of the ALK kinase domain. In this study, we have characterized activation loop mutations within the context of the full length ALK employing cell culture and Drosophila melanogaster model systems. Our results show that the Y1278S mutant observed in patients with neuroblastoma harbors gain-of-function activity. Secondly, we show that the suggested interaction between Y1278 and other amino acids might be of less importance in the activation process of the ALK kinase than previously proposed. Thirdly, of the three individual tyrosines in the 1278-YRASYY-1283 activation loop, we find that Y1283 is the critical tyrosine in the activation process. Taken together, our observations employing different model systems reveal new mechanistic insights on how the full length ALK receptor is activated and highlight differences with earlier described activation mechanisms observed in the NPM-ALK fusion protein, supporting a mechanism of activation more in line with those observed for the Insulin Receptor (InR).
Collapse
|
857
|
Abstract
Neuroblastoma is a cancer of the neural crest almost exclusively seen in childhood. While children with single, small primary tumors are often cured with surgery alone, the 65% of children with neuroblastoma whose disease has metastasized have less than a 50% chance of surviving five years after diagnosis. Innovative pharmacological strategies are critically needed for these children. Efforts to identify novel targets that afford ablation of neuroblastoma with minimal toxicity to normal tissues are underway. Developing approaches to neuroblastoma include those that target the catecholamine transporter, ubiquitin E3 ligase, the ganglioside GD2, the retinoic acid receptor, the protein kinases ALK and Aurora, and protein arginine N-methyltransferases. Here, as examples of the use of chemistry to combat neuroblastoma, we describe targeting of the protein arginine N-methyltransferases and their role in prolonging the half-life of the neuroblastoma oncoprotein N-Myc, redox signaling in neuroblastoma, and developmentally regulated proteins expressed in primitive neuroblastoma cells but not in mature neural crest elements.
Collapse
Affiliation(s)
- Jeanne N Hansen
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry , Rochester, New York 14642, United States
| | - Xingguo Li
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry , Rochester, New York 14642, United States
| | - Y George Zheng
- Department of Pharmaceutical and Biochemical Sciences, University of Georgia , Athens, Georgia 30602, United States
| | - Louis T Lotta
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry , Rochester, New York 14642, United States
| | - Abhishek Dedhe
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry , Rochester, New York 14642, United States
| | - Nina F Schor
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry , Rochester, New York 14642, United States
| |
Collapse
|
858
|
Zebrafish as a model to study neuroblastoma development. Cell Tissue Res 2017; 372:223-232. [PMID: 29027617 DOI: 10.1007/s00441-017-2702-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/11/2017] [Indexed: 02/08/2023]
Abstract
Neuroblastoma is a pediatric solid tumor arising from embryonic neural crest progenitor cells that normally generate the peripheral sympathetic nervous system. As such, the location of neuroblastoma tumors is correlated with the distribution of major post-ganglionic clusters throughout the sympathetic chain, with the highest incidence in the adrenal medulla or lumbar sympathetic ganglia (~65%). Neuroblastoma is an enigmatic tumor that can spontaneously regress with minimal treatment or become highly metastatic and develop resistance to aggressive treatments, including radiation and high-dose chemotherapy. Age of diagnosis, stage of disease and cellular and genetic features often predict whether the tumor will regress or advance to metastatic disease. Recent efforts using molecular and genomic technologies have allowed more accurate stratification of patients into low-, intermediate- and high-risk categories, thereby allowing for minimal intervention in low-risk patients and providing potential new therapeutic targets, such as the ALK receptor tyrosine kinase, for high-risk or relapsed patients. Despite these advances, the overall survival of high-risk neuroblastoma patients is still less than 50%. Furthermore, next-generation sequencing has revealed that almost two-thirds of neuroblastoma tumors do not contain obvious pathogenic mutations, suggesting that epigenetic mechanisms and/or a perturbed cellular microenvironment may heavily influence neuroblastoma development. Understanding the mechanisms that drive neuroblastoma, therefore, will likely require a combination of genomic, developmental and cancer biology approaches in whole animal systems. In this review, we discuss the contributions of zebrafish research to our understanding of neuroblastoma pathogenesis as well as the potential for this model system to accelerate the identification of more effective therapies for high-risk neuroblastoma patients in the future.
Collapse
|
859
|
Abstract
Neuroblastoma, an embryonal cancer of neural crest origin, shows metastases frequently at diagnosis. In this issue of Cancer Cell, Delloye-Bourgeois and colleagues demonstrate that neuroblastoma cell lines and patient-derived xenografts engraft and adopt a metastatic program in chick embryos. They identify Sema3C as a candidate switch that regulates metastatic spread.
Collapse
Affiliation(s)
- Tina Zheng
- Departments of Neurology, Pediatrics, and Neurosurgery, Brain Tumor Research Center, University of California, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - Marie Ménard
- Departments of Neurology, Pediatrics, and Neurosurgery, Brain Tumor Research Center, University of California, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| | - William A Weiss
- Departments of Neurology, Pediatrics, and Neurosurgery, Brain Tumor Research Center, University of California, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
860
|
Rifatbegovic F, Frech C, Abbasi MR, Taschner-Mandl S, Weiss T, Schmidt WM, Schmidt I, Ladenstein R, Ambros IM, Ambros PF. Neuroblastoma cells undergo transcriptomic alterations upon dissemination into the bone marrow and subsequent tumor progression. Int J Cancer 2017; 142:297-307. [PMID: 28921546 PMCID: PMC5725737 DOI: 10.1002/ijc.31053] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/28/2017] [Accepted: 08/02/2017] [Indexed: 12/12/2022]
Abstract
Neuroblastoma is the most common extracranial solid tumor in childhood. The vast majority of metastatic (M) stage patients present with disseminated tumor cells (DTCs) in the bone marrow (BM) at diagnosis and relapse. Although these cells represent a major obstacle in the treatment of neuroblastoma patients, insights into their expression profile remained elusive. The present RNA‐Seq study of stage 4/M primary tumors, enriched BM‐derived diagnostic and relapse DTCs, as well as the corresponding BM‐derived mononuclear cells (MNCs) from 53 patients revealed 322 differentially expressed genes in DTCs as compared to the tumors (q < 0.001, |log2FC|>2). Particularly, the levels of transcripts encoded by mitochondrial DNA were elevated in DTCs, whereas, for example, genes involved in angiogenesis were downregulated. Furthermore, 224 genes were highly expressed in DTCs and only slightly, if at all, in MNCs (q < 8 × 10−75 log2FC > 6). Interestingly, we found the transcriptome of relapse DTCs largely resembling those of diagnostic DTCs with only 113 differentially expressed genes under relaxed cut‐offs (q < 0.01, |log2FC|>0.5). Notably, relapse DTCs showed a positional enrichment of 31 downregulated genes on chromosome 19, including five tumor suppressor genes: SIRT6, BBC3/PUMA, STK11, CADM4 and GLTSCR2. This first RNA‐Seq analysis of neuroblastoma DTCs revealed their unique expression profile in comparison to the tumors and MNCs, and less pronounced differences between diagnostic and relapse DTCs. The latter preferentially affected downregulation of genes encoded by chromosome 19. As these alterations might be associated with treatment failure and disease relapse, further functional studies on DTCs should be considered. What's new? More than 90% of patients diagnosed with stage 4 metastatic (4/M) neuroblastoma present with disseminated tumor cells (DTCs) in the bone marrow (BM). Despite treatment, a substantial fraction of these patients experience disease relapse. Here, sequencing analysis of tumor tissue, BM‐derived mononuclear cells (MNCs), and DTCs from stage 4/M neuroblastoma patients indicates that numerous genes are differentially expressed in DTCs but are not or are only slightly altered in tumors and MNCs. Moreover, DTCs exhibited significant downregulation of tumor suppressor genes specifically on chromosome 19. Further studies are needed to determine whether DTC transcriptomic alterations are associated with neuroblastoma relapse.
Collapse
Affiliation(s)
- Fikret Rifatbegovic
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Christian Frech
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - M Reza Abbasi
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Sabine Taschner-Mandl
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Tamara Weiss
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Wolfgang M Schmidt
- Neuromuscular Research Department, Medical University of Vienna, Vienna, Austria
| | - Iris Schmidt
- Neuromuscular Research Department, Medical University of Vienna, Vienna, Austria
| | - Ruth Ladenstein
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Inge M Ambros
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Peter F Ambros
- Department of Tumor Biology, Children's Cancer Research Institute (CCRI), Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
861
|
Villasante A, Sakaguchi K, Kim J, Cheung N, Nakayama M, Parsa H, Okano T, Shimizu T, Vunjak-Novakovic G. Vascularized Tissue-Engineered Model for Studying Drug Resistance in Neuroblastoma. Am J Cancer Res 2017; 7:4099-4117. [PMID: 29158813 PMCID: PMC5695000 DOI: 10.7150/thno.20730] [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: 04/25/2017] [Accepted: 08/11/2017] [Indexed: 01/26/2023] Open
Abstract
Neuroblastoma is a vascularized pediatric tumor derived from neural crest stem cells that displays vasculogenic mimicry and can express a number of stemness markers, such as SOX2 and NANOG. Tumor relapse is the major cause of succumbing to this disease, and properties attributed to cancer stem-like cells (CSLC), such as drug-resistance and cell plasticity, seem to be the key mechanisms. However, the lack of controllable models that recapitulate the features of human neuroblastoma limits our understanding of the process and impedes the development of new therapies. In response to these limitations, we engineered a perfusable, vascularized in vitro model of three-dimensional human neuroblastoma to study the effects of retinoid therapy on tumor vasculature and drug-resistance. METHODS The in vitro model of neuroblastoma was generated using cell-sheet engineering and cultured in a perfusion bioreactor. Firstly, we stacked three cell sheets containing SKNBE(2) neuroblastoma cells and HUVEC. Then, a vascular bed made of fibrin, collagen I and HUVEC cells was placed onto a collagen-gel base with 8 microchannels. After gelling, the stacked cell sheets were placed on the vascular bed and cultured in the perfusion bioreactor (perfusion rate: 0.5 mL/min) for 4 days. Neuroblastoma models were treated with 10μM isotretionin in single daily doses for 5 days. RESULTS The bioengineered model recapitulated vasculogenic mimicry (vessel-like structure formation and tumor-derived endothelial cells-TECs), and contained CSLC expressing SOX2 and NANOG. Treatment with Isotretinoin destabilized vascular networks but failed to target vasculogenic mimicry and augmented populations of CSLCs expressing high levels of SOX2. Our results suggest that CSLCs can transdifferentiate into drug resistant CD31+-TECs, and reveal the presence of an intermediate state STEC (stem tumor-derived endothelial cell) expressing both SOX2 and CD31. CONCLUSION Our results reveal some roles of SOX2 in drug resistance and tumor relapse, and suggest that SOX2 could be a therapeutic target in neuroblastoma.
Collapse
|
862
|
Coughlan D, Gianferante M, Lynch CF, Stevens JL, Harlan LC. Treatment and survival of childhood neuroblastoma: Evidence from a population-based study in the United States. Pediatr Hematol Oncol 2017; 34:320-330. [PMID: 29039999 PMCID: PMC6764456 DOI: 10.1080/08880018.2017.1373315] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/26/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Childhood neuroblastoma describes a heterogeneous group of extracranial solid tumors, that are treated per risk profile. We sought to describe treatment patterns and survival using population-based data from throughout the United States. MATERIALS AND METHODS Using the National Cancer Institute (NCI)'s Patterns of Care data, we analyzed treatment provided to newly diagnosed, histologically confirmed neuroblastoma patients in 2010 and 2011, registered to one of 14 Surveillance, Epidemiology, and End Results (SEER) cancer registries. Data were re-abstracted from hospital records and treating physicians contacted for verification. Application of the Children's Oncology Group (COG)'s 3-level (low, intermediate and high) neuroblastoma risk classification system for therapeutic decision-making provided insight to community-based treatment patterns. Kaplan-Meier survival analyses, based on 5-years of follow-up, were also performed. RESULTS 76% of the 250 patients were enrolled on an open/active clinical trial. All low-risk patients received surgery. Most intermediate-risk patients (81%) received a chemotherapy regimen that included carboplatin, etoposide, cyclophosphamide and doxorubicin. High-risk patients received extensive, multimodal treatment consisting of chemotherapy, surgery, myeloablative chemotherapy with stem cell rescue (transplant), radiation, immunotherapy (dinutuximab), and isotretinoin therapy. 21% patients had died at the end of the maximum 60-month follow-up period. The 5-year estimated survival rates were lower for patients diagnosed with stage 4 disease, unfavorable DNA ploidy, MYCN gene amplification or classified as high-risk. CONCLUSION Most neuroblastoma patients are registered on a risk-based open/active clinical trial. Variation in modality, systemic agents and sequence of treatment reflects the heterogeneity of therapy received by these patients.
Collapse
Affiliation(s)
- Dairmuid Coughlan
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland 20892
| | - Matthew Gianferante
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Charles F Lynch
- University of Iowa College of Public Health, Iowa City, Iowa
| | | | - Linda C Harlan
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland 20892
| |
Collapse
|
863
|
Tümmler C, Snapkov I, Wickström M, Moens U, Ljungblad L, Maria Elfman LH, Winberg JO, Kogner P, Johnsen JI, Sveinbjørnsson B. Inhibition of chemerin/CMKLR1 axis in neuroblastoma cells reduces clonogenicity and cell viability in vitro and impairs tumor growth in vivo. Oncotarget 2017; 8:95135-95151. [PMID: 29221117 PMCID: PMC5707011 DOI: 10.18632/oncotarget.19619] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 07/06/2017] [Indexed: 02/06/2023] Open
Abstract
Pro-inflammatory cells, cytokines, and chemokines are essential in promoting a tumor supporting microenvironment. Chemerin is a chemotactic protein and a natural ligand for the receptors CMKLR1, GPR1, and CCRL2. The chemerin/CMKLR1 axis is involved in immunity and inflammation, and it has also been implicated in obesity and cancer. In neuroblastoma, a childhood tumor of the peripheral nervous system we identified correlations between high CMKLR1 and GPR1 expression and reduced overall survival probability. CMKLR1, GPR1, and chemerin RNA and protein were detected in neuroblastoma cell lines and neuroblastoma primary tumor tissue. Chemerin induced calcium mobilization, increased MMP-2 synthesis as well as MAP-kinase- and Akt-mediated signaling in neuroblastoma cells. Stimulation of neuroblastoma cells with serum, TNFα or IL-1β increased chemerin secretion. The small molecule CMKLR1 antagonist α-NETA reduced the clonogenicity and viability of neuroblastoma cell lines indicating the chemerin/CMKLR1 axis as a promoting factor in neuroblastoma tumorigenesis. Furthermore, nude mice carrying neuroblastoma SK-N-AS cells as xenografts showed impaired tumor growth when treated daily with α-NETA from day 1 after tumor cell injection. This study demonstrates the potential of the chemerin/CMKLR1 axis as a prognostic factor and possible therapeutic target in neuroblastoma.
Collapse
Affiliation(s)
- Conny Tümmler
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Science, University of Tromsø, Tromsø, Norway
| | - Igor Snapkov
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Science, University of Tromsø, Tromsø, Norway
| | - Malin Wickström
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ugo Moens
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Science, University of Tromsø, Tromsø, Norway
| | - Linda Ljungblad
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Lotta Helena Maria Elfman
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Jan-Olof Winberg
- Tumor Biology Research Group, Department of Medical Biology, Faculty of Health Science, University of Tromsø, Tromsø, Norway
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Baldur Sveinbjørnsson
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Science, University of Tromsø, Tromsø, Norway.,Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
864
|
Heterogeneity of neuroblastoma cell identity defined by transcriptional circuitries. Nat Genet 2017; 49:1408-1413. [PMID: 28740262 DOI: 10.1038/ng.3921] [Citation(s) in RCA: 302] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 06/28/2017] [Indexed: 02/06/2023]
Abstract
Neuroblastoma is a tumor of the peripheral sympathetic nervous system, derived from multipotent neural crest cells (NCCs). To define core regulatory circuitries (CRCs) controlling the gene expression program of neuroblastoma, we established and analyzed the neuroblastoma super-enhancer landscape. We discovered three types of identity in neuroblastoma cell lines: a sympathetic noradrenergic identity, defined by a CRC module including the PHOX2B, HAND2 and GATA3 transcription factors (TFs); an NCC-like identity, driven by a CRC module containing AP-1 TFs; and a mixed type, further deconvoluted at the single-cell level. Treatment of the mixed type with chemotherapeutic agents resulted in enrichment of NCC-like cells. The noradrenergic module was validated by ChIP-seq. Functional studies demonstrated dependency of neuroblastoma with noradrenergic identity on PHOX2B, evocative of lineage addiction. Most neuroblastoma primary tumors express TFs from the noradrenergic and NCC-like modules. Our data demonstrate a previously unknown aspect of tumor heterogeneity relevant for neuroblastoma treatment strategies.
Collapse
|
865
|
Van Roy N, Van Der Linden M, Menten B, Dheedene A, Vandeputte C, Van Dorpe J, Laureys G, Renard M, Sante T, Lammens T, De Wilde B, Speleman F, De Preter K. Shallow Whole Genome Sequencing on Circulating Cell-Free DNA Allows Reliable Noninvasive Copy-Number Profiling in Neuroblastoma Patients. Clin Cancer Res 2017; 23:6305-6314. [PMID: 28710315 DOI: 10.1158/1078-0432.ccr-17-0675] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/02/2017] [Accepted: 07/10/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Neuroblastoma (NB) is a heterogeneous disease characterized by distinct clinical features and by the presence of typical copy-number alterations (CNAs). Given the strong association of these CNA profiles with prognosis, analysis of the CNA profile at diagnosis is mandatory. Therefore, we tested whether the analysis of circulating cell-free DNA (cfDNA) present in plasma samples of patients with NB could offer a valuable alternative to primary tumor DNA for CNA profiling.Experimental Design: In 37 patients with NB, cfDNA analysis using shallow whole genome sequencing (sWGS) was compared with arrayCGH analysis of primary tumor tissue.Results: Comparison of CNA profiles on cfDNA showed highly concordant patterns, particularly in high-stage patients. Numerical chromosome imbalances as well as large and focal structural aberrations including MYCN and LIN28B amplification and ATRX deletion could be readily detected with sWGS using a low input of cfDNA.Conclusions: In conclusion, sWGS analysis on cfDNA offers a cost-effective, noninvasive, rapid, robust and sensitive alternative for tumor DNA copy-number profiling in most patients with NB. Clin Cancer Res; 23(20); 6305-14. ©2017 AACR.
Collapse
Affiliation(s)
- Nadine Van Roy
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Malaïka Van Der Linden
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Department of Pathology, Ghent University, Ghent, Belgium
| | - Björn Menten
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | | | - Charlotte Vandeputte
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University, Ghent, Belgium
| | - Geneviève Laureys
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Marleen Renard
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Leuven University Hospital, Leuven, Belgium
| | - Tom Sante
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Tim Lammens
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Bram De Wilde
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Frank Speleman
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Katleen De Preter
- Center for Medical Genetics, Ghent University, Ghent, Belgium. .,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| |
Collapse
|
866
|
Lodrini M, Sprüssel A, Astrahantseff K, Tiburtius D, Konschak R, Lode HN, Fischer M, Keilholz U, Eggert A, Deubzer HE. Using droplet digital PCR to analyze MYCN and ALK copy number in plasma from patients with neuroblastoma. Oncotarget 2017; 8:85234-85251. [PMID: 29156716 PMCID: PMC5689606 DOI: 10.18632/oncotarget.19076] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/20/2017] [Indexed: 01/28/2023] Open
Abstract
The invasive nature of surgical biopsies deters sequential application, and single biopsies often fail to reflect tumor dynamics, intratumor heterogeneity and drug sensitivities likely to change during tumor evolution and treatment. Implementing molecular characterization of cell-free neuroblastoma-derived DNA isolated from blood plasma could improve disease assessment for treatment selection and monitoring of patients with high-risk neuroblastoma. We established droplet digital PCR (ddPCR) protocols for MYCN and ALK copy number status in plasma from neuroblastoma patients. Our ddPCR protocol accurately discriminated between MYCN and ALK amplification, gain and normal diploid status in a large panel of neuroblastoma cell lines, and discrepancies with reported MYCN and ALK status were detected, including a high-level MYCN amplification in NB-1, a MYCN gain in SH-SY5Y, a high-level ALK amplification in IMR-32 and ALK gains in BE(2)-C, Kelly, SH-SY5Y and LAN-6. MYCN and ALK status were also reliably determined from cell-free DNA derived from medium conditioned by the cell lines. MYCN and ALK copy numbers of subcutaneous neuroblastoma xenograft tumors were accurately determined from cell-free DNA in the mouse blood plasma. In a final validation step, we accurately distinguished MYCN and ALK copy numbers of the corresponding primary tumors using retrospectively collected blood plasma samples from 10 neuroblastoma patients. Our data justify the further development of molecular disease characterization using cell-free DNA in blood plasma from patients with neuroblastoma. This expanded molecular diagnostic palette may improve monitoring of disease progression including relapse and metastatic events as well as therapy success or failure in high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Marco Lodrini
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Annika Sprüssel
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Daniela Tiburtius
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Konschak
- Translational Radiation Oncology Research Laboratory, Department of Radiooncology and Radiotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany
| | - Holger N Lode
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, Germany
| | - Matthias Fischer
- Department of Pediatric Hematology and Oncology, University Hospital Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Hedwig E Deubzer
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Junior Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC), Berlin, Germany
| |
Collapse
|
867
|
Mlakar V, Jurkovic Mlakar S, Lopez G, Maris JM, Ansari M, Gumy-Pause F. 11q deletion in neuroblastoma: a review of biological and clinical implications. Mol Cancer 2017; 16:114. [PMID: 28662712 PMCID: PMC5492892 DOI: 10.1186/s12943-017-0686-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/25/2017] [Indexed: 12/12/2022] Open
Abstract
Deletion of the long arm of chromosome 11 (11q deletion) is one of the most frequent events that occur during the development of aggressive neuroblastoma. Clinically, 11q deletion is associated with higher disease stage and decreased survival probability. During the last 25 years, extensive efforts have been invested to identify the precise frequency of 11q aberrations in neuroblastoma, the recurrently involved genes, and to understand the molecular mechanisms of 11q deletion, but definitive answers are still unclear. In this review, it is our intent to compile and review the evidence acquired to date on 11q deletion in neuroblastoma.
Collapse
Affiliation(s)
- Vid Mlakar
- CANSEARCH Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Simona Jurkovic Mlakar
- CANSEARCH Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205, Geneva, Switzerland
| | - Gonzalo Lopez
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - John M Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Marc Ansari
- CANSEARCH Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205, Geneva, Switzerland.,Department of Pediatrics, Onco-Hematology Unit, Geneva University Hospitals, Rue Willy-Donzé 6, 1205, Geneva, Switzerland
| | - Fabienne Gumy-Pause
- CANSEARCH Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205, Geneva, Switzerland. .,Department of Pediatrics, Onco-Hematology Unit, Geneva University Hospitals, Rue Willy-Donzé 6, 1205, Geneva, Switzerland.
| |
Collapse
|
868
|
Affiliation(s)
- Shoma Tsubota
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
869
|
Liu D, Tian S, Zhang K, Xiong W, Lubaki NM, Chen Z, Han W. Chimeric antigen receptor (CAR)-modified natural killer cell-based immunotherapy and immunological synapse formation in cancer and HIV. Protein Cell 2017; 8:861-877. [PMID: 28488245 PMCID: PMC5712291 DOI: 10.1007/s13238-017-0415-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/22/2017] [Indexed: 12/31/2022] Open
Abstract
Cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells contribute to the body’s immune defenses. Current chimeric antigen receptor (CAR)-modified T cell immunotherapy shows strong promise for treating various cancers and infectious diseases. Although CAR-modified NK cell immunotherapy is rapidly gaining attention, its clinical applications are mainly focused on preclinical investigations using the NK92 cell line. Despite recent advances in CAR-modified T cell immunotherapy, cost and severe toxicity have hindered its widespread use. To alleviate these disadvantages of CAR-modified T cell immunotherapy, additional cytotoxic cell-mediated immunotherapies are urgently needed. The unique biology of NK cells allows them to serve as a safe, effective, alternative immunotherapeutic strategy to CAR-modified T cells in the clinic. While the fundamental mechanisms underlying the cytotoxicity and side effects of CAR-modified T and NK cell immunotherapies remain poorly understood, the formation of the immunological synapse (IS) between CAR-modified T or NK cells and their susceptible target cells is known to be essential. The role of the IS in CAR T and NK cell immunotherapies will allow scientists to harness the power of CAR-modified T and NK cells to treat cancer and infectious diseases. In this review, we highlight the potential applications of CAR-modified NK cells to treat cancer and human immunodeficiency virus (HIV), and discuss the challenges and possible future directions of CAR-modified NK cell immunotherapy, as well as the importance of understanding the molecular mechanisms of CAR-modified T cell- or NK cell-mediated cytotoxicity and side effects, with a focus on the CAR-modified NK cell IS.
Collapse
Affiliation(s)
- Dongfang Liu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA. .,Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA.
| | - Shuo Tian
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Kai Zhang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Wei Xiong
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Ndongala Michel Lubaki
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Zhiying Chen
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Weidong Han
- Institute of Basic Medicine, College of Life Sciences, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
870
|
Xu Y, Chen X, Lin L, Chen H, Yu S, Li D. MicroRNA-149 is associated with clinical outcome in human neuroblastoma and modulates cancer cell proliferation through Rap1 independent of MYCN amplification. Biochimie 2017; 139:1-8. [PMID: 28456710 DOI: 10.1016/j.biochi.2017.04.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 04/24/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE We evaluated the clinical relevance of microRNA-149 (miR-149) in neuroblastoma (NB) and its functional roles in regulating NB proliferation in vitro. METHODS QRT-PCR was used to evaluate miR-149 expression in NB cell lines and primary NB tumors. Association between endogenous miR-149 expression in primary NB tumors and their host patients' clinicopathological factors and overall survival (OS) were statistically evaluated. In SH-SY5Y, an MYCN-non-amplified, and LAN5, an MYCN-amplified NB cell lines, miR-149 was either upregulated or downregulated by lentiviral transduction, to evaluate its effect on NB proliferation in vitro. Possible downstream target of miR-149, Ras-related protein 1 (Rap1), was evaluated by qRT-PCR and western blot in lentiviral-transduced NB cells. Moreover, Rap1 was either upregulated or downregulated in lentiviral-transduced NB cells to further evaluate its effect on miR-149-mediated NB proliferation in vitro. RESULTS MiR-149 is markedly downregulated in both in vitro NB cell lines and in vivo NB primary tumors. Low miR-149 expression is predominantly associated with Stage 3 or 4 primary NB tumors, and poor OS among NB patients. In SH-SY5Y and LAN5 cells, lentivirus-induced miR-149 upregulation inhibited, whereas miR-149 downregulation promoted NB proliferation in vitro, despite MYCN-amplification status. Rap1 expression, at both mRNA and protein levels, was inversely associated with miR-149 in NB. In addition, Rap1 upregulation or downregulation reversely regulated miR-149-mediated NB proliferation in vitro. CONCLUSION MiR-149 is downregulated in NB and closely associated with NB patients' clinical outcome. MiR-149 also functionally modulates NB cell proliferation in vitro, possibly through inverse-regulation on Rap1.
Collapse
Affiliation(s)
- Yali Xu
- Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350005, China
| | - Xinghe Chen
- Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350005, China
| | - Li Lin
- Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350005, China
| | - Hong Chen
- Office of Graduate Studies, Fujian Medical University, Fuzhou, Fujian Province, 350004, China
| | - Shuping Yu
- Office of Graduate Studies, Fujian Medical University, Fuzhou, Fujian Province, 350004, China
| | - Dumiao Li
- Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350005, China.
| |
Collapse
|
871
|
The MYCN Protein in Health and Disease. Genes (Basel) 2017; 8:genes8040113. [PMID: 28358317 PMCID: PMC5406860 DOI: 10.3390/genes8040113] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022] Open
Abstract
MYCN is a member of the MYC family of proto-oncogenes. It encodes a transcription factor, MYCN, involved in the control of fundamental processes during embryonal development. The MYCN protein is situated downstream of several signaling pathways promoting cell growth, proliferation and metabolism of progenitor cells in different developing organs and tissues. Conversely, deregulated MYCN signaling supports the development of several different tumors, mainly with a childhood onset, including neuroblastoma, medulloblastoma, rhabdomyosarcoma and Wilms’ tumor, but it is also associated with some cancers occurring during adulthood such as prostate and lung cancer. In neuroblastoma, MYCN-amplification is the most consistent genetic aberration associated with poor prognosis and treatment failure. Targeting MYCN has been proposed as a therapeutic strategy for the treatment of these tumors and great efforts have allowed the development of direct and indirect MYCN inhibitors with potential clinical use.
Collapse
|
872
|
Applebaum MA, Desai AV, Glade Bender JL, Cohn SL. Emerging and investigational therapies for neuroblastoma. Expert Opin Orphan Drugs 2017; 5:355-368. [PMID: 29062613 PMCID: PMC5649635 DOI: 10.1080/21678707.2017.1304212] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/06/2017] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Treatment for children with clinically aggressive, high-risk neuroblastoma remains challenging. Less than 50% of patients with high-risk neuroblastoma will survive long-term with current therapies, and survivors are at risk for serious treatment-related late toxicities. Here, we review new and evolving treatments that may ultimately improve outcome for children with high-risk neuroblastoma with decreased potential for late adverse events. AREAS COVERED New strategies for treating high-risk neuroblastoma are reviewed including: radiotherapy, targeted cytotoxics, biologics, immunotherapy, and molecularly targeted agents. Recently completed and ongoing neuroblastoma clinical trials testing these novel treatments are highlighted. In addition, we discuss ongoing clinical trials designed to evaluate precision medicine approaches that target actionable somatic mutations and oncogenic cellular pathways. EXPERT OPINION Advances in genomic medicine and molecular biology have led to the development of early phase studies testing biologically rational therapies targeting aberrantly activated cellular pathways. Because many of these drugs have a wider therapeutic index than standard chemotherapeutic agents, these treatments may be more effective and less toxic than current strategies. However, to effectively integrate these targeted strategies, robust predictive biomarkers must be developed that will identify patients who will benefit from these approaches and rapidly match treatments to patients at diagnosis.
Collapse
Affiliation(s)
- Mark A. Applebaum
- Department of Pediatrics, University of Chicago, Chicago, Illinois, 60637, United States of America
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois, 60637, United States of America
| | - Ami V. Desai
- Department of Pediatrics, University of Chicago, Chicago, Illinois, 60637, United States of America
| | - Julia L. Glade Bender
- Department of Pediatrics, Columbia University Medical Center, New York, New York, 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, 10032
| | - Susan L. Cohn
- Department of Pediatrics, University of Chicago, Chicago, Illinois, 60637, United States of America
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois, 60637, United States of America
| |
Collapse
|
873
|
Gaze MN. Semi-quantitative scoring of skeletal metastases by 123I-mIBG scintigraphy in high-risk neuroblastoma. Eur J Nucl Med Mol Imaging 2017; 44:1251-1253. [PMID: 28258443 DOI: 10.1007/s00259-017-3660-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 02/17/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Mark N Gaze
- Department of Oncology, University College London Hospitals NHS Foundation Trust, 250 Euston Road, London, NW1 2PG, UK.
| |
Collapse
|
874
|
Abbasi MR, Rifatbegovic F, Brunner C, Mann G, Ziegler A, Pötschger U, Crazzolara R, Ussowicz M, Benesch M, Ebetsberger-Dachs G, Chan GCF, Jones N, Ladenstein R, Ambros IM, Ambros PF. Impact of Disseminated Neuroblastoma Cells on the Identification of the Relapse-Seeding Clone. Clin Cancer Res 2017; 23:4224-4232. [PMID: 28228384 DOI: 10.1158/1078-0432.ccr-16-2082] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/20/2016] [Accepted: 02/12/2017] [Indexed: 02/07/2023]
Abstract
Purpose: Tumor relapse is the most frequent cause of death in stage 4 neuroblastomas. Since genomic information on the relapse precursor cells could guide targeted therapy, our aim was to find the most appropriate tissue for identifying relapse-seeding clones.Experimental design: We analyzed 10 geographically and temporally separated samples of a single patient by SNP array and validated the data in 154 stage 4 patients.Results: In the case study, aberrations unique to certain tissues and time points were evident besides concordant aberrations shared by all samples. Diagnostic bone marrow-derived disseminated tumor cells (DTCs) as well as the metastatic tumor and DTCs at relapse displayed a 1q deletion, not detected in any of the seven primary tumor samples. In the validation cohort, the frequency of 1q deletion was 17.8%, 10%, and 27.5% in the diagnostic DTCs, diagnostic tumors, and DTCs at relapse, respectively. This aberration was significantly associated with 19q and ATRX deletions. We observed a significant increased likelihood of an adverse event in the presence of 19q deletion in the diagnostic DTCs.Conclusions: Different frequencies of 1q and 19q deletions in the primary tumors as compared with DTCs, their relatively high frequency at relapse, and their effect on event-free survival (19q deletion) indicate the relevance of analyzing diagnostic DTCs. Our data support the hypothesis of a branched clonal evolution and a parallel progression of primary and metastatic tumor cells. Therefore, searching for biomarkers to identify the relapse-seeding clone should involve diagnostic DTCs alongside the tumor tissue. Clin Cancer Res; 23(15); 4224-32. ©2017 AACR.
Collapse
Affiliation(s)
- M Reza Abbasi
- CCRI, Children's Cancer Research Institute, Vienna, Austria.
| | | | | | - Georg Mann
- St. Anna Children's Hospital, Vienna, Austria
| | - Andrea Ziegler
- CCRI, Children's Cancer Research Institute, Vienna, Austria
| | | | - Roman Crazzolara
- Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Marek Ussowicz
- Department of Pediatric Hematology and Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Martin Benesch
- Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Graz, Austria
| | | | - Godfrey C F Chan
- Department of Pediatrics and Adolescent Medicine, University of Hong Kong, Hong Kong
| | - Neil Jones
- Department of Pediatrics and Adolescent Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Ruth Ladenstein
- CCRI, Children's Cancer Research Institute, Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Inge M Ambros
- CCRI, Children's Cancer Research Institute, Vienna, Austria
| | - Peter F Ambros
- CCRI, Children's Cancer Research Institute, Vienna, Austria. .,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|