151
|
Abstract
Most children who succumb to solid malignancies do so because of the burden of metastatic disease or due to complications associated with the therapy administered to treat metastatic disease. Approximately one-quarter of children with solid tumors will present with metastatic disease, and an additional 20% ultimately develop metastatic disease, most commonly in the lung. The role of surgery in the treatment of metastatic solid tumors, given its disseminated nature, is not intuitive, yet there are circumstances in which surgical resection of metastatic disease can potentially be curative. However, the utility of surgery is very much dependent on histology, and generally is most appropriate for those malignancies with histologies that are refractory to other adjuvant therapies.
Collapse
Affiliation(s)
- Todd E. Heaton
- Pediatric Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew M. Davidoff
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
152
|
Kalaskar RR, Kalaskar AR. Neuroblastoma in early childhood: A rare case report and review of literature. Contemp Clin Dent 2016; 7:401-4. [PMID: 27630510 PMCID: PMC5004559 DOI: 10.4103/0976-237x.188579] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuroblastoma is an extremely rare pediatric neoplasm whose prognosis becomes poor and poor as the age advances. It can be sporadic or nonfamilial in origin. It is primarily a tumor of abdominal origin from where it metastasis to lymph nodes, liver, intracranial and orbital sites, and central nervous system. There is no standard dental treatment protocol for the management of neuroblastoma due to its poor survival rate and rarity. However, dental treatment may follow the protocol of preventive and restorative. Surgicals should be performed under supervision as it may trigger metastasis. We report a rare case of neuroblastoma in a 3-year-old child presenting classical oral manifestations such as bilateral palatal swelling, rolled border ulcer on the posterior part of hard palate adjacent to primary molars, and bilateral proptosis.
Collapse
Affiliation(s)
- Ritesh R Kalaskar
- Department of Pedodontics, Government Dental College and Hospital, Nagpur, Maharashtra, India
| | - Ashita R Kalaskar
- Department of Oral Diagnosis Medicine and Radiology, VSPM Dental College and Research Center, Nagpur, Maharashtra, India
| |
Collapse
|
153
|
Morgenstern DA, London WB, Stephens D, Volchenboum SL, Simon T, Nakagawara A, Shimada H, Schleiermacher G, Matthay KK, Cohn SL, Pearson AD, Irwin MS. Prognostic significance of pattern and burden of metastatic disease in patients with stage 4 neuroblastoma: A study from the International Neuroblastoma Risk Group database. Eur J Cancer 2016; 65:1-10. [DOI: 10.1016/j.ejca.2016.06.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 05/08/2016] [Accepted: 06/08/2016] [Indexed: 01/09/2023]
|
154
|
Norozi F, Ahmadzadeh A, Shahrabi S, Vosoughi T, Saki N. Mesenchymal stem cells as a double-edged sword in suppression or progression of solid tumor cells. Tumour Biol 2016; 37:11679-11689. [PMID: 27440203 DOI: 10.1007/s13277-016-5187-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/13/2016] [Indexed: 02/07/2023] Open
Abstract
Tumor cells are able to attract mesenchymal stem cells (MSCs) to primary tumor site. On the other hand, MSCs secrete various factors to attract tumor cells towards BM. In this review, in addition to assessment of MSCs function at tumor sites and their impact on growth and metastasis of tumor cells, the importance of MSC in attraction of malignant cells to BM and their involvement in drug resistance of tumor cells have also been studied. Relevant literature was identified by a PubMed search (2000-2015) of English-language literature using the terms mesenchymal stem cells, cancer cell, metastasis, and tumor microenvironment. MSCs migrate towards tumor microenvironment and are involved in both pro-tumorigenic and antitumorigenic functions. The dual function of MSCs at tumor sites is dependent upon a variety of factors, including the type and origin of MSCs, the cancer cell line under study, in vivo or in vitro conditions, the factors secreted by MSCs and interactions between MSCs, host immune cells and cancer cells. Therefore, MSCs can be regarded both as friends and enemies of cancer cells. Although the role of a number of pathways, including IL-6/STAT3 pathway, has been indicated in controlling the interaction between MSCs and tumor cells, other mechanisms by which MSCs can control the tumor cells are not clear yet. A better understanding of these mechanisms through further studies can determine the exact role of MSCs in cancer progression and identify them as important therapeutic agents or targets.
Collapse
Affiliation(s)
- Fatemeh Norozi
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ahmad Ahmadzadeh
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of biochemistry and hematology, Faculty of Medicine, Semnan University of medical sciences, Semnan, Iran
| | - Tina Vosoughi
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
155
|
Lau DT, Flemming CL, Gherardi S, Perini G, Oberthuer A, Fischer M, Juraeva D, Brors B, Xue C, Norris MD, Marshall GM, Haber M, Fletcher JI, Ashton LJ. MYCN amplification confers enhanced folate dependence and methotrexate sensitivity in neuroblastoma. Oncotarget 2016; 6:15510-23. [PMID: 25860940 PMCID: PMC4558167 DOI: 10.18632/oncotarget.3732] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/10/2015] [Indexed: 12/12/2022] Open
Abstract
MYCN amplification occurs in 20% of neuroblastomas and is strongly related to poor clinical outcome. We have identified folate-mediated one-carbon metabolism as highly upregulated in neuroblastoma tumors with MYCN amplification and have validated this finding experimentally by showing that MYCN amplified neuroblastoma cell lines have a higher requirement for folate and are significantly more sensitive to the antifolate methotrexate than cell lines without MYCN amplification. We have demonstrated that methotrexate uptake in neuroblastoma cells is mediated principally by the reduced folate carrier (RFC; SLC19A1), that SLC19A1 and MYCN expression are highly correlated in both patient tumors and cell lines, and that SLC19A1 is a direct transcriptional target of N-Myc. Finally, we assessed the relationship between SLC19A1 expression and patient survival in two independent primary tumor cohorts and found that SLC19A1 expression was associated with increased risk of relapse or death, and that SLC19A1 expression retained prognostic significance independent of age, disease stage and MYCN amplification. This study adds upregulation of folate-mediated one-carbon metabolism to the known consequences of MYCN amplification, and suggests that this pathway might be targeted in poor outcome tumors with MYCN amplification and high SLC19A1 expression.
Collapse
Affiliation(s)
- Diana T Lau
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Claudia L Flemming
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | | | - Giovanni Perini
- Department of Biology, University of Bologna, Bologna, Italy
| | - André Oberthuer
- Children's Hospital, Department of Pediatric Oncology and Hematology, University of Cologne and Centre for Molecular Medicine Cologne, Cologne, Germany
| | - Matthias Fischer
- Children's Hospital, Department of Pediatric Oncology and Hematology, University of Cologne and Centre for Molecular Medicine Cologne, Cologne, Germany
| | - Dilafruz Juraeva
- Division of Theoretical Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Benedikt Brors
- Division of Theoretical Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Chengyuan Xue
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Murray D Norris
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Glenn M Marshall
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, Randwick, NSW, Australia
| | - Lesley J Ashton
- Faculty of Medicine, School of Women's and Children's Health, University of New South Wales, Sydney, NSW, Australia.,Research Portfolio, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
156
|
Černý I, Prášek J, Kašpárková H. Superiority of SPECT/CT over planar 123I-mIBG images in neuroblastoma patients with impact on Curie and SIOPEN score values. Nuklearmedizin 2016; 55:151-7. [PMID: 27054367 DOI: 10.3413/nukmed-0743-15-05] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 03/11/2016] [Indexed: 11/20/2022]
Abstract
OBJECTIVE The existing most common semi-quantitative systems used for neuroblastoma diagnosis include Curie and SIOPEN scores, which are based on 123I-MIBG planar scans. The purpose of our study was to find out whether a statistically significant difference exists in evaluation based on planar and SPECT/CT scans. We also compared the Curie and SIOPEN methods in terms of their use in regular practice. Patients; method: 45 patients aged 0-10 years; 213 assessments were done in total, and the Curie and SIOPEN scores were determined in each case based on planar and SPECT/CT scans. Student's T-test and the Bland-Altman plot were used for the statistical analysis. RESULTS Both methods demonstrated a statistically significant difference (p < 0.0001) between planar and SPECT/CT evaluation. In the group of 35 patients with neuroblastoma in clinical stages 3 and 4, in 54% of the patients SPECT/CT detected a lesion that was not visible in the planar scan. In 89% of cases, the lesion was confirmed by another imaging method (CT, MRI). In the group of 10 patients in the clinical stage 1, a difference between planar and SPECT/CT scanning was found only in one patient (10%). In the whole set, 25% patients showed a pathological finding only in soft tissues. CONCLUSION We recommend to perform semiquantitative evaluation of neuroblastoma based on SPECT/CT scans, particularly in patients in clinical stages 3 and 4. It is advisable to include soft tissues in the score assessment, as well, given that only soft tissues may be involved in up to 25.
Collapse
Affiliation(s)
- Igor Černý
- Igor Černý, KNM FN Brno, Jihlavská 20, 62500 Brno, Czech Republic, Tel. +420/777277427, Fax +420/532233840,
| | | | | |
Collapse
|
157
|
Harreld JH, Bratton EM, Federico SM, Li X, Grover W, Li Y, Kerr NC, Wilson MW, Hoehn ME. Orbital Metastasis Is Associated With Decreased Survival in Stage M Neuroblastoma. Pediatr Blood Cancer 2016; 63:627-33. [PMID: 26599346 PMCID: PMC5127440 DOI: 10.1002/pbc.25847] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/21/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Approximately 30% of patients with metastatic (stage M) neuroblastoma present with periorbital ecchymosis from orbital osseous disease. Though locoregional disease is staged by imaging, the prognostic significance of metastatic site in stage M disease is unknown. We hypothesize that, compared to nonorbital metastasis, orbital metastasis is associated with decreased survival in patients with stage M neuroblastoma, and that periorbital ecchymosis reflects location and extent of orbital disease. PROCEDURE Medical records and imaging from 222 patients with stage M neuroblastoma seen at St. Jude Children's Research Hospital between January 1995 and May 2009 were reviewed. Thirty-seven patients were <18 months of age at diagnosis and 185 were ≥18 months of age. Overall survival (OS) and 5-year survival (5YS) were compared for patients with and without orbital, calvarial and nonorbital osseous metastasis, and with and without periorbital ecchymosis (log-rank test). Associations of periorbital ecchymosis with orbital metastasis location/extent were explored (Fisher's exact test, t-test). RESULTS In patients ≥18 months of age, only orbital metastasis was associated with decreased 5YS (P = 0.0323) and OS (P = 0.0288). In patients <18 months of age, neither orbital, calvarial, or nonorbital bone metastasis was associated with OS or 5YS. Periorbital ecchymosis was associated with higher number of involved orbital bones (P = 0.0135), but not location or survival. CONCLUSIONS In patients ≥ 18 months of age with stage M neuroblastoma, orbital metastatic disease is associated with decreased 5YS and OS. In future clinical trials, orbital disease may be useful as an imaging-based risk factor for substratification of stage M neuroblastoma.
Collapse
Affiliation(s)
- Julie H. Harreld
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN
| | | | - Sara M. Federico
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Xingyu Li
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - William Grover
- Department of Ophthalmology, Medstar Georgetown University Hospital, Washington, DC
| | - Yimei Li
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Natalie C Kerr
- Department of Ophthalmology, University of TN Hamilton Eye Institute, Memphis, TN; Department of Pediatrics, University of TN College of Medicine, Memphis, TN; Division of Ophthalmology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Matthew W. Wilson
- Department of Ophthalmology, University of TN Hamilton Eye Institute, Memphis, TN; Division of Ophthalmology, St. Jude Children’s Research Hospital, Memphis, TN, Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Mary E. Hoehn
- Department of Ophthalmology, University of TN Hamilton Eye Institute, Memphis, TN; Department of Pediatrics, University of TN College of Medicine, Memphis, TN; Division of Ophthalmology, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
158
|
Smith JR, Moreno L, Heaton SP, Chesler L, Pearson ADJ, Garrett MD. Novel pharmacodynamic biomarkers for MYCN protein and PI3K/AKT/mTOR pathway signaling in children with neuroblastoma. Mol Oncol 2016; 10:538-52. [PMID: 26686971 PMCID: PMC5423144 DOI: 10.1016/j.molonc.2015.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/11/2015] [Indexed: 11/22/2022] Open
Abstract
There is an urgent need for improved therapies for children with high-risk neuroblastoma where survival rates remain low. MYCN amplification is the most common genomic change associated with aggressive neuroblastoma and drugs targeting PI3K/AKT/mTOR, to activate MYCN oncoprotein degradation, are entering clinical evaluation. Our aim was to develop and validate pharmacodynamic (PD) biomarkers to evaluate both proof of mechanism and proof of concept for drugs that block PI3K/AKT/mTOR pathway activity in children with neuroblastoma. We have addressed the issue of limited access to tumor biopsies for quantitative detection of protein biomarkers by optimizing a three-color fluorescence activated cell sorting (FACS) method to purify CD45-/GD2+/CD56+ neuroblastoma cells from bone marrow. We then developed a novel quantitative measurement of MYCN protein in these isolated neuroblastoma cells, providing the potential to demonstrate proof of concept for drugs that inhibit PI3K/AKT/mTOR signaling in this disease. In addition we have established quantitative detection of three biomarkers for AKT pathway activity (phosphorylated and total AKT, GSK3β and P70S6K) in surrogate platelet-rich plasma (PRP) from pediatric patients. Together our new approach to neuroblastoma cell isolation for protein detection and suite of PD assays provides for the first time the opportunity for robust, quantitative measurement of protein-based PD biomarkers in this pediatric patient population. These will be ideal tools to support clinical evaluation of PI3K/AKT/mTOR pathway drugs and their ability to target MYCN oncoprotein in upcoming clinical trials in neuroblastoma.
Collapse
Affiliation(s)
- Jennifer R Smith
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, United Kingdom; Division of Clinical Studies, The Institute of Cancer Research, London, SM2 5NG, United Kingdom
| | - Lucas Moreno
- Division of Clinical Studies, The Institute of Cancer Research, London, SM2 5NG, United Kingdom; Children's and Young People's Unit, Royal Marsden NHS Foundation Trust, Sutton, SM2 5PT, United Kingdom; CNIO, Spanish National Cancer Research Centre, Melchor Fernandez Almagro 3, 28029, Madrid, Spain
| | - Simon P Heaton
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, United Kingdom
| | - Louis Chesler
- Division of Clinical Studies, The Institute of Cancer Research, London, SM2 5NG, United Kingdom; Children's and Young People's Unit, Royal Marsden NHS Foundation Trust, Sutton, SM2 5PT, United Kingdom
| | - Andrew D J Pearson
- Division of Clinical Studies, The Institute of Cancer Research, London, SM2 5NG, United Kingdom; Children's and Young People's Unit, Royal Marsden NHS Foundation Trust, Sutton, SM2 5PT, United Kingdom
| | - Michelle D Garrett
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, United Kingdom; School of Biosciences, University of Kent, Canterbury, Kent, CT2 7NJ, United Kingdom.
| |
Collapse
|
159
|
Morrison MA, Zimmerman MW, Look AT, Stewart RA. Studying the peripheral sympathetic nervous system and neuroblastoma in zebrafish. Methods Cell Biol 2016; 134:97-138. [PMID: 27312492 DOI: 10.1016/bs.mcb.2015.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The zebrafish serves as an excellent model to study vertebrate development and disease. Optically clear embryos, combined with tissue-specific fluorescent reporters, permit direct visualization and measurement of peripheral nervous system formation in real time. Additionally, the model is amenable to rapid cellular, molecular, and genetic approaches to determine how developmental mechanisms contribute to disease states, such as cancer. In this chapter, we describe the development of the peripheral sympathetic nervous system (PSNS) in general, and our current understanding of genetic pathways important in zebrafish PSNS development specifically. We also illustrate how zebrafish genetics is used to identify new mechanisms controlling PSNS development and methods for interrogating the potential role of PSNS developmental pathways in neuroblastoma pathogenesis in vivo using the zebrafish MYCN-driven neuroblastoma model.
Collapse
Affiliation(s)
- M A Morrison
- University of Utah, Salt Lake City, UT, United States
| | | | - A T Look
- Harvard Medical School, Boston, MA, United States
| | - R A Stewart
- University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
160
|
Middelbeek J, Visser D, Henneman L, Kamermans A, Kuipers AJ, Hoogerbrugge PM, Jalink K, van Leeuwen FN. TRPM7 maintains progenitor-like features of neuroblastoma cells: implications for metastasis formation. Oncotarget 2016; 6:8760-76. [PMID: 25797249 PMCID: PMC4496182 DOI: 10.18632/oncotarget.3315] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/08/2015] [Indexed: 12/18/2022] Open
Abstract
Neuroblastoma is an embryonal tumor derived from poorly differentiated neural crest cells. Current research is aimed at identifying the molecular mechanisms that maintain the progenitor state of neuroblastoma cells and to develop novel therapeutic strategies that induce neuroblastoma cell differentiation. Mechanisms controlling neural crest development are typically dysregulated during neuroblastoma progression, and provide an appealing starting point for drug target discovery. Transcriptional programs involved in neural crest development act as a context dependent gene regulatory network. In addition to BMP, Wnt and Notch signaling, activation of developmental gene expression programs depends on the physical characteristics of the tissue microenvironment. TRPM7, a mechanically regulated TRP channel with kinase activity, was previously found essential for embryogenesis and the maintenance of undifferentiated neural crest progenitors. Hence, we hypothesized that TRPM7 may preserve progenitor-like, metastatic features of neuroblastoma cells. Using multiple neuroblastoma cell models, we demonstrate that TRPM7 expression closely associates with the migratory and metastatic properties of neuroblastoma cells in vitro and in vivo. Moreover, microarray-based expression profiling on control and TRPM7 shRNA transduced neuroblastoma cells indicates that TRPM7 controls a developmental transcriptional program involving the transcription factor SNAI2. Overall, our data indicate that TRPM7 contributes to neuroblastoma progression by maintaining progenitor-like features.
Collapse
Affiliation(s)
- Jeroen Middelbeek
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Daan Visser
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Linda Henneman
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Arthur J Kuipers
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Peter M Hoogerbrugge
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Princes Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Kees Jalink
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Frank N van Leeuwen
- Laboratory of Pediatric Oncology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
161
|
Sivakumar S, Poulik J, Sivaswamy L. Monocular Blindness as Presentation Manifestation of Neuroblastoma. Neurohospitalist 2016; 6:41. [PMID: 26740857 DOI: 10.1177/1941874415585546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Sanjeev Sivakumar
- Department of Neurology, Detroit Medical Center/Wayne State University, Detroit, MI, USA
| | - Janet Poulik
- Department of Pathology, Children's Hospital of Michigan, Detroit, MI, USA
| | - Lalitha Sivaswamy
- Department of Pediatrics and Neurology, Children's Hospital of Michigan, Detroit, MI, USA
| |
Collapse
|
162
|
Forster JI, Köglsberger S, Trefois C, Boyd O, Baumuratov AS, Buck L, Balling R, Antony PMA. Characterization of Differentiated SH-SY5Y as Neuronal Screening Model Reveals Increased Oxidative Vulnerability. ACTA ACUST UNITED AC 2016; 21:496-509. [PMID: 26738520 PMCID: PMC4904349 DOI: 10.1177/1087057115625190] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/10/2015] [Indexed: 02/01/2023]
Abstract
The immortalized and proliferative cell line SH-SY5Y is one of the most commonly used cell lines in neuroscience and neuroblastoma research. However, undifferentiated SH-SY5Y cells share few properties with mature neurons. In this study, we present an optimized neuronal differentiation protocol for SH-SY5Y that requires only two work steps and 6 days. After differentiation, the cells present increased levels of ATP and plasma membrane activity but reduced expression of energetic stress response genes. Differentiation results in reduced mitochondrial membrane potential and decreased robustness toward perturbations with 6-hydroxydopamine. We are convinced that the presented differentiation method will leverage genetic and chemical high-throughput screening projects targeting pathways that are involved in the selective vulnerability of neurons with high energetic stress levels.
Collapse
Affiliation(s)
- J I Forster
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - S Köglsberger
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - C Trefois
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - O Boyd
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - A S Baumuratov
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - L Buck
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - R Balling
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| | - P M A Antony
- Luxembourg Centre for Systems Biomedicine (LCSB), Esch-sur-Alzette, Luxembourg
| |
Collapse
|
163
|
Left Ventricular Metastasis in Neuroblastoma: A Case Report. J Pediatr Hematol Oncol 2016; 38:74-7. [PMID: 26479997 DOI: 10.1097/mph.0000000000000439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Neuroblastoma is the most common extracranial solid tumor in children. Most common sites of metastases from neuroblastoma are bone marrow, bone and lymph nodes, however cardiac metastasis is rarely seen. Metastatic cardiac tumors are 20 to 40 times more common than primary cardiac tumors. Mechanism of cardiac metastasis can be hematogenous, lymphatogenous, and direct extension/infiltration of tumor cells. Usually right heart metastasis is seen. Left ventricular metastatic tumor has never been reported with neuroblastoma.
Collapse
|
164
|
Garner EF, Beierle EA. Cancer Stem Cells and Their Interaction with the Tumor Microenvironment in Neuroblastoma. Cancers (Basel) 2015; 8:cancers8010005. [PMID: 26729169 PMCID: PMC4728452 DOI: 10.3390/cancers8010005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 12/16/2015] [Accepted: 12/21/2015] [Indexed: 12/26/2022] Open
Abstract
Neuroblastoma, a solid tumor arising from neural crest cells, accounts for over 15% of all pediatric cancer deaths. The interaction of neuroblastoma cancer-initiating cells with their microenvironment likely plays an integral role in the maintenance of resistant disease and tumor relapse. In this review, we discuss the interaction between neuroblastoma cancer-initiating cells and the elements of the tumor microenvironment and how these interactions may provide novel therapeutic targets for this difficult to treat disease.
Collapse
Affiliation(s)
- Evan F Garner
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, AL 35233, USA.
| | - Elizabeth A Beierle
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, AL 35233, USA.
| |
Collapse
|
165
|
Borriello L, Seeger RC, Asgharzadeh S, DeClerck YA. More than the genes, the tumor microenvironment in neuroblastoma. Cancer Lett 2015; 380:304-14. [PMID: 26597947 DOI: 10.1016/j.canlet.2015.11.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/06/2015] [Accepted: 11/11/2015] [Indexed: 10/22/2022]
Abstract
Neuroblastoma is the second most common solid tumor in children. Since the seminal discovery of the role of amplification of the MYCN oncogene in the pathogenesis of neuroblastoma in the 1980s, much focus has been on the contribution of genetic alterations in the progression of this cancer. However it is now clear that not only genetic events play a role but that the tumor microenvironment (TME) substantially contributes to the biology of neuroblastoma. In this article, we present a comprehensive review of the literature on the contribution of the TME to the ten hallmarks of cancer in neuroblastoma and discuss the mechanisms of communication between neuroblastoma cells and the TME that underlie the influence of the TME on neuroblastoma progression. We end our review by discussing how the knowledge acquired over the last two decades in this field is now leading to new clinical trials targeting the TME.
Collapse
Affiliation(s)
- Lucia Borriello
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA; Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Robert C Seeger
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA; Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Shahab Asgharzadeh
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA; Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Yves A DeClerck
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA; Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA; Department of Biochemistry and Molecular Biology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
166
|
Vandenhaute E, Stump-Guthier C, Lasierra Losada M, Tenenbaum T, Rudolph H, Ishikawa H, Schwerk C, Schroten H, Dürken M, März M, Karremann M. The choroid plexus may be an underestimated site of tumor invasion to the brain: an in vitro study using neuroblastoma cell lines. Cancer Cell Int 2015; 15:102. [PMID: 26500454 PMCID: PMC4619509 DOI: 10.1186/s12935-015-0257-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/14/2015] [Indexed: 01/14/2023] Open
Abstract
Background The central nervous system (CNS) is protected by several barriers, including the blood–brain (BBB) and blood-cerebrospinal fluid (BCSFB) barriers. Understanding how cancer cells circumvent these protective barriers to invade the CNS is of crucial interest, since brain metastasis during cancer is often a fatal event in both children and adults. However, whereas much effort has been invested in elucidating the process of tumor cell transmigration across the BBB, the role of the BCSFB might still be underestimated considering the significant number of meningeal cancer involvement. Our work aimed to investigate the transmigration of neuroblastoma cells across the BCSFB in vitro. Methods We used an inverted model of the human BCSFB presenting proper restrictive features including adequate expression of tight-junction proteins, low permeability to integrity markers, and high trans-epithelial electrical resistance. Two different human neuroblastoma cell lines (SH-SY5Y and IMR-32) were used to study the transmigration process by fluorescent microscopy analysis. Results The results show that neuroblastoma cells are able to actively cross the tight human in vitro BCSFB model within 24 h. The presence and transmigration of neuroblastoma cancer cells did not affect the barrier integrity within the duration of the experiment. Conclusions In conclusion, we presume that the choroid plexus might be an underestimated site of CNS invasion, since neuroblastoma cell lines are able to actively cross a choroid plexus epithelial cell layer. Further studies are warranted to elucidate the molecular mechanisms of tumor cell transmigration in vitro and in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12935-015-0257-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elodie Vandenhaute
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), EA 2465, Université d'Artois (UArtois), Lens, France
| | - Carolin Stump-Guthier
- Department of Pediatric and Adolescent Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - María Lasierra Losada
- Institute of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Tobias Tenenbaum
- Department of Pediatric and Adolescent Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Henriette Rudolph
- Department of Pediatric and Adolescent Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hiroshi Ishikawa
- Department of NDU Life Sciences, School of Life Dentistry, The Nippon Dental University, Tokyo, Japan
| | - Christian Schwerk
- Department of Pediatric and Adolescent Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Horst Schroten
- Department of Pediatric and Adolescent Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias Dürken
- Department of Pediatric and Adolescent Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Martin März
- Department of Pediatric and Adolescent Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Karremann
- Department of Pediatric and Adolescent Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
167
|
Honokiol induces autophagy of neuroblastoma cells through activating the PI3K/Akt/mTOR and endoplasmic reticular stress/ERK1/2 signaling pathways and suppressing cell migration. Cancer Lett 2015; 370:66-77. [PMID: 26454217 DOI: 10.1016/j.canlet.2015.08.030] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 08/23/2015] [Accepted: 08/25/2015] [Indexed: 11/20/2022]
Abstract
In children, neuroblastomas are the most common and deadly solid tumor. Our previous study showed that honokiol, a small-molecule polyphenol, can traverse the blood-brain barrier and kill neuroblastoma cells. In this study, we further investigated the mechanisms of honokiol-induced insults to neuroblastoma cells. Treatment of neuroblastoma neuro-2a cells with honokiol elevated the levels of microtubule-associated protein light chain 3 (LC3)-II and induced cell autophagy in time- and concentration-dependent manners. Interestingly, pretreatment with 3-methyladenine (3-MA), an inhibitor of autophagy, led to the simultaneous attenuation of honokiol-induced cell autophagy and apoptosis but did not influence cell necrosis. As to the mechanisms, exposure of neuro-2a cells to honokiol time-dependently decreased the amount of phosphatidylinositol 3-kinase (PI3K). Sequentially, honokiol downregulated phosphorylation of protein kinase B (Akt) and mammalian target of rapamycin (mTOR) in neuro-2a cells. Furthermore, honokiol elevated the levels of glucose-regulated protein (GpR)78, an endoplasmic reticular stress (ERS)-associated protein, and amounts of intracellular reactive oxygen species (ROS). In contrast, reducing production of intracellular ROS using N-acetylcysteine, a scavenger of ROS, concurrently suppressed honokiol-induced cellular autophagy. Consequently, honokiol stimulated phosphorylation of extracellular signal-regulated kinase (ERK)1/2. However, pretreatment of neuro-2a cells with PD98059, an inhibitor of ERK1/2, lowered honokiol-induced autophagy. The effects of honokiol on inducing autophagy and apoptosis of neuroblastoma cells were further confirmed using mouse neuroblastoma NB41A3 cells as our experimental model. Fascinatingly, treatment of neuroblastoma neuro-2a and NB41A3 cells with honokiol for 12 h did not affect cell autophagy or apoptosis but caused significant suppression of cell migration. Taken together, this study showed that honokiol can induce autophagy of neuroblastoma cells and consequent apoptosis through activating the PI3K/Akt/mTOR and ERS/ROS/ERK1/2 signaling pathways and suppressing cell migration. Thus, honokiol has potential for treating neuroblastomas.
Collapse
|
168
|
Lee JW, Cho A, Yun M, Lee JD, Lyu CJ, Kang WJ. Prognostic value of pretreatment FDG PET in pediatric neuroblastoma. Eur J Radiol 2015; 84:2633-9. [PMID: 26462795 DOI: 10.1016/j.ejrad.2015.09.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 09/21/2015] [Accepted: 09/27/2015] [Indexed: 11/26/2022]
Abstract
PURPOSE This study aimed to evaluate the prognostic value of pretreatment (18)F-fluorodeoxyglucose (FDG) positron emission tomography (PET) in pediatric neuroblastoma patients. METHODS The study included 50 pediatric neuroblastoma patients who underwent diagnostic work-up FDG PET before any treatment. The maximum standardized uptake value (SUV(max)) of the primary tumor lesion (P(max)), the SUV(max) of all the tumor lesions, including the primary tumor lesion and metastatic lesions (T(max)), and the uptake ratio of T(max) to mean SUV of normal liver tissue (T(max)/L(mean)) were calculated and tested as prognostic factors. RESULTS Of the 50 patients, 15 (30.0%) experienced disease progression and 21 (42.0%) died during the follow-up period. On univariate analysis, the histopathology, tumor stage, bone marrow involvement, serum levels of lactate dehydrogenase (LDH), neuron-specific enolase, and ferritin, primary tumor size, P(max), T(max), and T(max)/L(mean) were significant prognostic factors for disease progression-free survival (PFS), whereas the tumor stage, serum level of LDH, T(max), and T(max)/L(mean) were determined to be significant for predicting overall survival (OS). On multivariate analysis, the histopathology and serum level of LDH were independent prognostic factors for PFS, and only the T(max)/L(mean) was an independent prognostic factor for OS. The 2-year PFS and OS rates were over 80.0% in patients with low FDG uptake, meanwhile, patients with high FDG uptake showed the 2-year PFS of less than 30.0% and OS of less than 55.0%. CONCLUSION FDG PET was an independent prognostic factor for OS in neuroblastoma patients. FDG PET can provide effective information on the prognosis for neuroblastoma patients.
Collapse
Affiliation(s)
- Jeong Won Lee
- Department of Nuclear Medicine, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, 25 Simgok-ro 100 beon-gil, Seo-gu, Incheon 404-834, South Korea
| | - Arthur Cho
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemoon-gu, Seoul 120-752, South Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemoon-gu, Seoul 120-752, South Korea
| | - Jong Doo Lee
- Department of Radiology, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, 25 Simgok-ro 100 beon-gil, Seo-gu, 404-834, South Korea
| | - Chuhl Joo Lyu
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemoon-gu, Seoul 120-752, South Korea.
| | - Won Jun Kang
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemoon-gu, Seoul 120-752, South Korea.
| |
Collapse
|
169
|
Glass JJ, Phillips PA, Gunning PW, Stehn JR. Hypoxia alters the recruitment of tropomyosins into the actin stress fibres of neuroblastoma cells. BMC Cancer 2015; 15:712. [PMID: 26475688 PMCID: PMC4608101 DOI: 10.1186/s12885-015-1741-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/09/2015] [Indexed: 01/27/2023] Open
Abstract
Background Neuroblastoma is the most common extracranial solid tumor of childhood. The heterogeneous microenvironment of solid tumors contains hypoxic regions associated with poor prognosis and chemoresistance. Hypoxia implicates the actin cytoskeleton through its essential roles in motility, invasion and proliferation. However, hypoxia-induced changes in the actin cytoskeleton have only recently been observed in human cells. Tropomyosins are key regulators of the actin cytoskeleton and we hypothesized that tropomyosins may mediate hypoxic phenotypes. Methods Neuroblastoma (SH-EP) cells were incubated ± hypoxia (1 % O2, 5 % CO2) for up to 144 h, before examining the cytoskeleton by confocal microscopy and Western blotting. Results Hypoxic cells were characterized by a more organized actin cytoskeleton and a reduced ability to degrade gelatin substrates. Hypoxia significantly increased mean actin filament bundle width (72 h) and actin filament length (72–96 h). This correlated with increased hypoxic expression and filamentous organization of stabilizing tropomyosins Tm1 and Tm2. However, isoform specific changes in tropomyosin expression were more evident at 96 h. Conclusions This study demonstrates hypoxia-induced changes in the recruitment of high molecular weight tropomyosins into the actin stress fibres of a human cancer. While hypoxia induced clear changes in actin organization compared with parallel normoxic cultures of neuroblastoma, the precise role of tropomyosins in this hypoxic actin reorganization remains to be determined. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1741-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joshua J Glass
- Oncology Research Unit, School of Medical Sciences, UNSW Australia, Room 229, Wallace Wurth Building, Sydney, NSW, 2052, Australia. .,Current address: ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3010, Australia.
| | - Phoebe A Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, UNSW Australia, Sydney, NSW, 2052, Australia.
| | - Peter W Gunning
- Oncology Research Unit, School of Medical Sciences, UNSW Australia, Room 229, Wallace Wurth Building, Sydney, NSW, 2052, Australia.
| | - Justine R Stehn
- Oncology Research Unit, School of Medical Sciences, UNSW Australia, Room 229, Wallace Wurth Building, Sydney, NSW, 2052, Australia.
| |
Collapse
|
170
|
Parikh NS, Howard SC, Chantada G, Israels T, Khattab M, Alcasabas P, Lam CG, Faulkner L, Park JR, London WB, Matthay KK. SIOP-PODC adapted risk stratification and treatment guidelines: Recommendations for neuroblastoma in low- and middle-income settings. Pediatr Blood Cancer 2015; 62:1305-16. [PMID: 25810263 PMCID: PMC5132052 DOI: 10.1002/pbc.25501] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/30/2015] [Indexed: 12/11/2022]
Abstract
Neuroblastoma is the most common extracranial solid tumor in childhood in high-income countries (HIC), where consistent treatment approaches based on clinical and tumor biological risk stratification have steadily improved outcomes. However, in low- and middle- income countries (LMIC), suboptimal diagnosis, risk stratification, and treatment may occur due to limited resources and unavailable infrastructure. The clinical practice guidelines outlined in this manuscript are based on current published evidence and expert opinions. Standard risk stratification and treatment explicitly adapted to graduated resource settings can improve outcomes for children with neuroblastoma by reducing preventable toxic death and relapse.
Collapse
Affiliation(s)
- Nehal S. Parikh
- Department of PediatricsDivision of Hematology‐OncologyConnecticut Children's Medical CenterHartfordConnecticut
| | | | | | - Trijn Israels
- VU University Medical CenterAmsterdamthe Netherlands
| | - Mohammed Khattab
- Department of PaediatricsChildren's Hospital of RabatRabatMorocco
| | - Patricia Alcasabas
- University of the Philippines‐Philippine General HospitalManilaPhilippines
| | - Catherine G. Lam
- Department of Oncology and International Outreach ProgramSt. Jude Children's Research HospitalMemphisTennessee
| | | | - Julie R. Park
- Seattle Children's HospitalUniversity of Washington School of Medicine and Fred Hutchinson Cancer Research CenterSeattleWashington
| | - Wendy B. London
- Harvard Medical SchoolBoston Children's Hospital and Dana Farber Cancer InstituteBostonMaryland
| | - Katherine K. Matthay
- Department of PediatricsUCSF School of Medicine and UCSF Benioff Children's HospitalSan FranciscoCalifornia
| |
Collapse
|
171
|
Right adrenal gland neuroblastoma infiltrating the liver and mimicking mesenchymal hamartoma: A case report. Int J Surg Case Rep 2015; 12:95-8. [PMID: 26036461 PMCID: PMC4486106 DOI: 10.1016/j.ijscr.2015.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 05/08/2015] [Accepted: 05/19/2015] [Indexed: 11/29/2022] Open
Abstract
The right adrenal gland neuroblastoma and infiltrated the adjacent liver substance mimicking mesenchymal hamartoma of the liver. The Presentation of fever of unknown origin in a case of right adrenal gland neuroblastoma is rare. Neuroblastoma should be considered in differential diagnosis of abdominal mass in all infants and children.
Introduction Neuroblastoma is the most common extracranial solid pediatric malignancy. The most common site is abdomen with predominance of suprarenal medulla. Infiltration of the tumour to the liver is rare. No cases were reported in the literature about the misdiagnosis of neuroblastoma as mesenchymal hamartoma in the liver. Presentation of case We represent a rare case of neuroblastoma misdiagnosed as mesenchymal hamartoma in liver in a six-month-old female infant presented with fever and abdominal mass. Abdominal computed tomography (CT) revealed large cystic lesion occupying most of the right liver enchroaching upon right suprarenal region and displacing the right kidney inferior suggestive for mesenchymal hamartoma. Right adrenalectomy with en-bloc resection of the adjacent liver segments was done. Postoperative pathology revealed neuroblastoma with positive specific immunohistochemistry (IHC). Discussion Although neuroblastoma is the second most common pediatric abdominal malignancy with specific diagnostic modalities, a misdiagnosis of a case with neuroblastoma as mesenchymal hamartoma is rare. Histopathological diagnosis of neuroblastoma with positive IHC is essential as shown in our case. Conclusion We represent a rare case of neuroblastoma which arose from the right adrenal gland and infiltrated the adjacent liver substance mimicking mesenchymal hamartoma of the liver. Neuroblastoma is rarely presented with pyrexia of unknown origin. Neuroblastoma should be considered in differential diagnosis of abdominal mass in all infants and children.
Collapse
|
172
|
Federico SM, Brady SL, Pappo A, Wu J, Mao S, McPherson VJ, Young A, Furman WL, Kaufman R, Kaste S. The role of chest computed tomography (CT) as a surveillance tool in children with high-risk neuroblastoma. Pediatr Blood Cancer 2015; 62:976-81. [PMID: 25641708 PMCID: PMC4694045 DOI: 10.1002/pbc.25400] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 11/21/2014] [Indexed: 01/07/2023]
Abstract
BACKGROUND Standardization of imaging obtained in children with neuroblastoma is not well established. This study examines chest CT in pediatric patients with high-risk neuroblastoma. PROCEDURE Medical records and imaging from 88 patients with high-risk neuroblastoma, diagnosed at St. Jude Children's Research Hospital between January, 2002 and December, 2009, were reviewed. Surveillance imaging was conducted through 2013. Ten patients with thoracic disease at diagnosis were excluded. Event free survival (EFS) and overall survival (OS) were estimated. Size specific dose estimates for CT scans of the chest, abdomen, and pelvis were used to estimate absolute organ doses to 23 organs. Organ dosimetry was used to calculate cohort effective dose. RESULTS The 5 year OS and EFS were 51.9% ± 6.5% and 42.6% ± 6.5%, respectively. Forty-six (58.9%) patients progressed/recurred and 41 (52.6%) died of disease. Eleven patients (14%) developed thoracic disease progression/recurrence identified by chest CT (1 paraspinal mass, 1 pulmonary nodules, and 9 nodal). MIBG (metaiodobenzylguanidine) scans identified thoracic disease in six patients. Five of the 11 had normal chest MIBG scans; three were symptomatic and two were asymptomatic with normal chest MIBG scans but avid bone disease. The estimated radiation dose savings from surveillance without CT chest imaging was 42%, 34% when accounting for modern CT acquisition (2011-2013). CONCLUSIONS Neuroblastoma progression/recurrence in the chest is rare and often presents with symptoms or is identified using standard non-CT imaging modalities. For patients with non-thoracic high-risk neuroblastoma at diagnosis, omission of surveillance chest CT imaging can save 35-42% of the radiation burden without compromising disease detection.
Collapse
Affiliation(s)
- Sara M Federico
- Departments of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee; Departments of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Young SA, McCabe KE, Bartakova A, Delaney J, Pizzo DP, Newbury RO, Varner JA, Schlaepfer DD, Stupack DG. Integrin α4 Enhances Metastasis and May Be Associated with Poor Prognosis in MYCN-low Neuroblastoma. PLoS One 2015; 10:e0120815. [PMID: 25973900 PMCID: PMC4431816 DOI: 10.1371/journal.pone.0120815] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/08/2015] [Indexed: 12/11/2022] Open
Abstract
High-risk neuroblastoma is associated with an overall survival rate of 30–50%. Neuroblastoma-expressed cell adhesion receptors of the integrin family impact cell adhesion, migration, proliferation and survival. Integrin α4 is essential for neural crest cell motility during development, is highly expressed on leukocytes, and is critical for transendothelial migration. Thus, cancer cells that express this receptor may exhibit increased metastatic potential. We show that α4 expression in human and murine neuroblastoma cell lines selectively enhances in vitro interaction with the alternatively spliced connecting segment 1 of fibronectin, as well as vascular cell adhesion molecule-1 and increases migration. Integrin α4 expression enhanced experimental metastasis in a syngeneic tumor model, reconstituting a pattern of organ involvement similar to that seen in patients. Accordingly, antagonism of integrin α4 blocked metastasis, suggesting adhesive function of the integrin is required. However, adhesive function was not sufficient, as mutants of integrin α4 that conserved the matrix-adhesive and promigratory function in vitro were compromised in their metastatic capacity in vivo. Clinically, integrin α4 is more frequently expressed in non-MYNC amplified tumors, and is selectively associated with poor prognosis in this subset of disease. These results reveal an unexpected role for integrin α4 in neuroblastoma dissemination and identify α4 as a potential prognostic indicator and therapeutic target.
Collapse
Affiliation(s)
- Shanique A. Young
- Division of Gynecologic Oncology, Department of Reproductive Medicine, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, United States of America
- University of California San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, California, 92093, United States of America
| | - Katelyn E. McCabe
- Division of Gynecologic Oncology, Department of Reproductive Medicine, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, United States of America
- University of California San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, California, 92093, United States of America
| | - Alena Bartakova
- Division of Gynecologic Oncology, Department of Reproductive Medicine, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, United States of America
| | - Joe Delaney
- Division of Gynecologic Oncology, Department of Reproductive Medicine, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, United States of America
- University of California San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, California, 92093, United States of America
| | - Donald P. Pizzo
- University of California San Diego Center for Advanced Laboratory Medicine, 10300 Campus Point Drive, MC7210, Room 1253, San Diego, CA, 92121, United States of America
- Department of Pathology, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, United States of America
| | - Robert O. Newbury
- Department of Pathology, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, United States of America
| | - Judith A. Varner
- University of California San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, California, 92093, United States of America
- Department of Pathology, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, United States of America
| | - David D. Schlaepfer
- Division of Gynecologic Oncology, Department of Reproductive Medicine, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, United States of America
- University of California San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, California, 92093, United States of America
| | - Dwayne G. Stupack
- Division of Gynecologic Oncology, Department of Reproductive Medicine, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, California, 92093, United States of America
- University of California San Diego Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, California, 92093, United States of America
- * E-mail:
| |
Collapse
|
174
|
Huang YY, Tzen KY, Liu YL, Chiu CH, Tsai CL, Wen HP, Tang KH, Liu CC, Shiue CY. Impact of residual 18F-fluoride in 18F-FDOPA for the diagnosis of neuroblastoma. Ann Nucl Med 2015; 29:489-98. [DOI: 10.1007/s12149-015-0970-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/30/2015] [Indexed: 01/11/2023]
|
175
|
Abstract
In patients with high-risk metastatic neuroblastoma, the benefit of radiation therapy (RT) to metastatic sites as part of primary treatment has not been fully investigated. The purpose of this single-institution study was to evaluate local control of irradiated metastatic sites, and characterize metastatic disease burden and anatomic distribution in patients with high-risk metastatic neuroblastoma. The records of all patients diagnosed with stage 4 neuroblastoma between August 2000 and January 2010 were reviewed. Exclusion criteria included: bone-marrow only metastatic site, total body irradiation, or no imaging follow-up. A total of 37 patients met eligibility criteria. Median follow-up period for patients without relapse was 61 months. Five-year overall survival for all patients was 67%. Thirteen patients (35%) received RT to a metastatic site as part of their primary treatment. Among these patients, in-field recurrence occurred in three patients (23%), including two of three treated calvarial sites. In patients treated with or without RT to a metastatic site, respectively, there was no significant difference in 5-year overall survival (73% vs. 63%, P=0.84) or relapse-free survival (46% and 55%, P=0.48). Current metastatic site RT dose may be suboptimal, and certain locations may predict for a poor response. Further studies are necessary to elucidate the optimal role of RT to metastatic sites.
Collapse
|
176
|
Cussó L, Mirones I, Peña-Zalbidea S, García-Vázquez V, García-Castro J, Desco M. Combination of single-photon emission computed tomography and magnetic resonance imaging to track 111in-oxine-labeled human mesenchymal stem cells in neuroblastoma-bearing mice. Mol Imaging 2015; 13. [PMID: 25248941 DOI: 10.2310/7290.2014.00033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Homing is an inherent, complex, multistep process performed by cells such as human bone marrow mesenchymal stem cells (hMSCs) to travel from a distant location to inflamed or damaged tissue and tumors. This ability of hMSCs has been exploited as a tumor-targeting strategy in cell-based cancer therapy. The purpose of this study was to investigate the applicability of 111In-oxine for tracking hMSCs in vivo by combining single-photon emission computed tomography (SPECT) and magnetic resonance imaging (MRI). 111In-labeled hMSCs (106 cells) were infused intraperitoneally in neuroblastoma-bearing mice, whereas a control group received a dose of free 111In-oxine. SPECT and MRI studies were performed 24 and 48 hours afterwards. Initially, the images showed similar activity in the abdomen in both controls and hMSC-injected animals. In general, abdominal activity decreases at 48 hours. hMSC-injected animals showed increased uptake in the tumor area at 48 hours, whereas the control group showed a low level of activity at 24 hours, which decreased at 48 hours. In conclusion, tracking 111In-labeled hMSCs combining SPECT and MRI is feasible and may be transferable to clinical research. The multimodal combination is essential to ensure appropriate interpretation of the images.
Collapse
|
177
|
DuBois SG, Groshen S, Park JR, Haas-Kogan DA, Yang X, Geier E, Chen E, Giacomini K, Weiss B, Cohn SL, Granger MM, Yanik GA, Hawkins R, Courtier J, Jackson H, Goodarzian F, Shimada H, Czarnecki S, Tsao-Wei D, Villablanca JG, Marachelian A, Matthay KK. Phase I Study of Vorinostat as a Radiation Sensitizer with 131I-Metaiodobenzylguanidine (131I-MIBG) for Patients with Relapsed or Refractory Neuroblastoma. Clin Cancer Res 2015; 21:2715-21. [PMID: 25695691 DOI: 10.1158/1078-0432.ccr-14-3240] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 02/07/2015] [Indexed: 01/17/2023]
Abstract
PURPOSE (131)I-metaiodobenzylguanidine (MIBG) is a radiopharmaceutical with activity in neuroblastoma. Vorinostat is a histone deacetylase inhibitor that has radiosensitizing properties. The goal of this phase I study was to determine the MTDs of vorinostat and MIBG in combination. EXPERIMENTAL DESIGN Patients ≤ 30 years with relapsed/refractory MIBG-avid neuroblastoma were eligible. Patients received oral vorinostat (dose levels 180 and 230 mg/m(2)) daily days 1 to 14. MIBG (dose levels 8, 12, 15, and 18 mCi/kg) was given on day 3 and peripheral blood stem cells on day 17. Alternating dose escalation of vorinostat and MIBG was performed using a 3+3 design. RESULTS Twenty-seven patients enrolled to six dose levels, with 23 evaluable for dose escalation. No dose-limiting toxicities (DLT) were seen in the first three dose levels. At dose level 4 (15 mCi/kg MIBG/230 mg/m(2) vorinostat), 1 of 6 patients had DLT with grade 4 hypokalemia. At dose level 5 (18 mCi/kg MIBG/230 mg/m(2) vorinostat), 2 patients had dose-limiting bleeding (one grade 3 and one grade 5). At dose level 5a (18 mCi/kg MIBG/180 mg/m(2) vorinostat), 0 of 6 patients had DLT. The most common toxicities were neutropenia and thrombocytopenia. The response rate was 12% across all dose levels and 17% at dose level 5a. Histone acetylation increased from baseline in peripheral blood mononuclear cells collected on days 3 and 12 to 14. CONCLUSIONS Vorinostat at 180 mg/m(2)/dose is tolerable with 18 mCi/kg MIBG. A phase II trial comparing this regimen to single-agent MIBG is ongoing.
Collapse
Affiliation(s)
- Steven G DuBois
- Department of Pediatrics, University of California San Francisco, San Francisco, California.
| | - Susan Groshen
- Department of Preventive Medicine, USC Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, California
| | - Julie R Park
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Xiaodong Yang
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Ethan Geier
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California
| | - Eugene Chen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California
| | - Kathy Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California
| | - Brian Weiss
- Department of Pediatrics, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Susan L Cohn
- Department of Pediatrics, University of Chicago School of Medicine, Chicago, Illinois
| | - M Meaghan Granger
- Department of Pediatrics, Cook Children's Hospital, Fort Worth, Texas
| | - Gregory A Yanik
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Randall Hawkins
- Department of Radiology, University of California San Francisco, San Francisco, California
| | - Jesse Courtier
- Department of Radiology, University of California San Francisco, San Francisco, California
| | - Hollie Jackson
- Department of Radiology, USC Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, California
| | - Fariba Goodarzian
- Department of Radiology, USC Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, California
| | - Hiroyuki Shimada
- Department of Pathology, USC Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, California
| | - Scarlett Czarnecki
- Department of Pediatrics, USC Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, California
| | - Denice Tsao-Wei
- Department of Preventive Medicine, USC Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, California
| | - Judith G Villablanca
- Department of Pediatrics, USC Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, California
| | - Araz Marachelian
- Department of Pediatrics, USC Keck School of Medicine and Children's Hospital Los Angeles, Los Angeles, California
| | - Katherine K Matthay
- Department of Pediatrics, University of California San Francisco, San Francisco, California
| |
Collapse
|
178
|
Uslu L, Donig J, Link M, Rosenberg J, Quon A, Daldrup-Link HE. Value of 18F-FDG PET and PET/CT for evaluation of pediatric malignancies. J Nucl Med 2015; 56:274-86. [PMID: 25572088 DOI: 10.2967/jnumed.114.146290] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Successful management of solid tumors in children requires imaging tests for accurate disease detection, characterization, and treatment monitoring. Technologic developments aim toward the creation of integrated imaging approaches that provide a comprehensive diagnosis with a single visit. These integrated diagnostic tests not only are convenient for young patients but also save direct and indirect health-care costs by streamlining procedures, minimizing hospitalizations, and minimizing lost school or work time for children and their parents. (18)F-FDG PET/CT is a highly sensitive and specific imaging modality for whole-body evaluation of pediatric malignancies. However, recent concerns about ionizing radiation exposure have led to a search for alternative imaging methods, such as whole-body MR imaging and PET/MR. As we develop new approaches for tumor staging, it is important to understand current benchmarks. This review article will synthesize the current literature on (18)F-FDG PET/CT for tumor staging in children, summarizing questions that have been solved and providing an outlook on unsolved avenues.
Collapse
Affiliation(s)
- Lebriz Uslu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California; and
| | - Jessica Donig
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California; and
| | - Michael Link
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Jarrett Rosenberg
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California; and
| | - Andrew Quon
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California; and
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, California; and
| |
Collapse
|
179
|
Isolated late CNS relapse in a young adult 10 years after initial treatment for neuroblastoma. J Pediatr Hematol Oncol 2015; 37:75-7. [PMID: 24755834 DOI: 10.1097/mph.0000000000000163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
180
|
Tsutsumimoto T, Williams P, Yoneda T. The SK-N-AS human neuroblastoma cell line develops osteolytic bone metastases with increased angiogenesis and COX-2 expression. J Bone Oncol 2014; 3:67-76. [PMID: 26909300 PMCID: PMC4723652 DOI: 10.1016/j.jbo.2014.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/03/2014] [Indexed: 12/17/2022] Open
Abstract
Neuroblastoma (NB), which arises from embryonic neural crest cells, is the most common extra-cranial solid tumor of childhood. Approximately half of NB patients manifest bone metastasis accompanied with bone pain, fractures and bone marrow failure, leading to disturbed quality of life and poor survival. To study the mechanism of bone metastasis of NB, we established an animal model in which intracardiac inoculation of the SK-N-AS human NB cells in nude mice developed osteolytic bone metastases with increased osteoclastogenesis. SK-N-AS cells induced the expression of receptor activator of NF-κB ligand and osteoclastogenesis in mouse bone marrow cells in the co-culture. SK-N-AS cells expressed COX-2 mRNA and produced substantial amounts of prostaglandin E2 (PGE2). In contrast, the SK-N-DZ and SK-N-FI human NB cells failed to develop bone metastases, induce osteoclastogenesis, express COX-2 mRNA and produce PGE2. Immunohistochemical examination of SK-N-AS bone metastasis and subcutaneous tumor showed strong expression of COX-2. The selective COX-2 inhibitor NS-398 inhibited PGE2 production and suppressed bone metastases with reduced osteoclastogenesis. NS-398 also inhibited subcutaneous SK-N-AS tumor development with decreased angiogenesis and vascular endothelial growth factor-A expression. Of interest, metastasis to the adrenal gland, a preferential site for NB development, was also diminished by NS-398. Our results suggest that COX2/PGE2 axis plays a critical role in the pathophysiology of osteolytic bone metastases and tumor development of the SK-NS-AS human NB. Inhibition of angiogenesis by suppressing COX-2/PGE2 may be an effective therapeutic approach for children with NB.
Collapse
Affiliation(s)
- Takahiro Tsutsumimoto
- Division of Endocrinology and Metabolism, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA; Department of Cellular and Molecular Biochemistry, Osaka University Graduate School of Dentistry, Suita, Osaka 565-0871, Japan
| | - Paul Williams
- Division of Endocrinology and Metabolism, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA
| | - Toshiyuki Yoneda
- Division of Endocrinology and Metabolism, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA; Department of Cellular and Molecular Biochemistry, Osaka University Graduate School of Dentistry, Suita, Osaka 565-0871, Japan
| |
Collapse
|
181
|
Kiyonari S, Kadomatsu K. Neuroblastoma models for insights into tumorigenesis and new therapies. Expert Opin Drug Discov 2014; 10:53-62. [DOI: 10.1517/17460441.2015.974544] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
182
|
Adult neuroblastoma complicated by increased intracranial pressure: a case report and review of the literature. Case Rep Oncol Med 2014; 2014:341980. [PMID: 25328733 PMCID: PMC4190830 DOI: 10.1155/2014/341980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/30/2014] [Accepted: 09/03/2014] [Indexed: 11/29/2022] Open
Abstract
Neuroblastoma is the third most commonly occurring malignancy of the pediatric population, although it is extremely rare in the adult population. In adults, neuroblastoma is often metastatic and portends an extremely poor overall survival. Our case report documents metastatic neuroblastoma occurring in a healthy 29-year-old woman whose course was complicated by an unusual presentation of elevated intracranial pressures. The patient was treated with systemic chemotherapy, I131 metaiodobenzylguanidine (MIBG) radiotherapy, and autologous stem cell transplant (SCT). Unfortunately the patient's response to therapy was limited and she subsequently died. We aim to review neuroblastoma in the context of increased intracranial pressure and the limited data of neuroblastoma occurring in the adult population, along with proposed treatment options.
Collapse
|
183
|
Morandi F, Corrias MV, Pistoia V. Evaluation of bone marrow as a metastatic site of human neuroblastoma. Ann N Y Acad Sci 2014; 1335:23-31. [PMID: 25315505 DOI: 10.1111/nyas.12554] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Arising from neural crest cells, neuroblastoma (NB) is the most common extracranial pediatric solid tumor. The clinical presentation of NB is heterogeneous, ranging from patients with asymptomatic tumor masses, who require minimal treatment, to patients with metastatic disease who are treated with multimodal therapies. Clinical outcome is also variable, with overall survival ranging from 98% to 100% in infants with stage 1 NB, to less than 30% in patients with stage 4 MYCN-amplified NB. More than 50% of patients show metastasis at diagnosis, with the involvement of different vascularized tissues, including the bone marrow (BM). In this paper, we focus on BM infiltration by NB cells, which is considered an adverse prognostic factor. In particular, we discuss the role of different biological factors that may favor the dissemination of NB cells in the BM, such as chromosomic abnormalities, gene amplification, transcription factors, cell-surface receptors, products of oncogenes, and, more importantly, cytokines and chemokines. In addition, we analyze different techniques to evaluate BM infiltration by malignant cells (i.e., flow cytometry, immunocytochemistry, and quantitative reverse transcriptase polymerase chain reaction). Finally, we review recent data regarding phenotypic and genetic characterization of BM-infiltrating malignant cells and characterization of the BM microenvironment in NB patients compared to healthy subjects.
Collapse
Affiliation(s)
- Fabio Morandi
- Laboratory of Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | | | | |
Collapse
|
184
|
Abstract
The overall prognosis for most pediatric cancers is good. Mortality for all childhood cancers combined is approximately half what it was in 1975, and the survival rates of many malignancies continue to improve. However, the incidence of childhood cancer is significant and the related emergencies that develop acutely carry significant morbidity and mortality. Emergency providers who can identify and manage oncologic emergencies can contribute significantly to an improved prognosis. Effective care of pediatric malignancies requires an age-appropriate approach to patients and compassionate understanding of family dynamics.
Collapse
Affiliation(s)
- Melanie K Prusakowski
- Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, 1906 Belleview Avenue, Roanoke, VA 24014, USA.
| | - Daniel Cannone
- Virginia Tech Carilion School of Medicine, 1906 Belleview Avenue, Roanoke, VA 24014, USA
| |
Collapse
|
185
|
Rogowitz E, Babiker HM, Kanaan M, Millius RA, Ringenberg QS, Bishop M. Neuroblastoma of the elderly, an oncologist's nightmare: case presentation, literature review and SEER database analysis. Exp Hematol Oncol 2014; 3:20. [PMID: 25071978 PMCID: PMC4112985 DOI: 10.1186/2162-3619-3-20] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/11/2014] [Indexed: 12/29/2022] Open
Abstract
Neuroblastoma is considered a pediatric malignancy as over 95% of cases are diagnosed in patients ≤10 years old. This cancer is extremely rare in elderly patients. We conducted a Surveillance, Epidemiology, and End Results (SEER) database analysis in the USA between 1973–2007 that revealed only 35 elderly patients (>60 years of age) with neuroblastoma of whom only 2 patients had primary mediastinal neuroblastoma. There is a paucity of treatment and survival outcomes data for the elderly owing to the rarity of neuroblastoma in this population. Currently there are no standard guidelines or protocols for treatment of adult neuroblastoma. We report a rare and challenging case of an 86-year old patient presenting with mediastinal neuroblastoma and syndrome of inappropriate antidiuretic hormone secretion (SIADH) successfully treated with resection. Herein, we also provide a review of the literature and updated survival data on neuroblastoma based on results of our SEER database review.
Collapse
Affiliation(s)
- Elisa Rogowitz
- University of Arizona College of Medicine, 1501 N. Campbell Ave, Tucson, AZ 85724, USA.,Division of Hematology-Oncology, Department of Medicine, Southern Arizona VA Health Care System, 3601 S 6Th Ave, Tucson, AZ 85723, USA
| | - Hani M Babiker
- Division of Hematology-Oncology, Department of Medicine, Southern Arizona VA Health Care System, 3601 S 6Th Ave, Tucson, AZ 85723, USA.,Division of Hematology-Oncology, Department of Medicine, University of Arizona College of Medicine, 1501 North Campbell Avenue, Tucson, AZ 85724, USA.,Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, AZ 85724, USA
| | - Mohammed Kanaan
- Division of Hematology-Oncology, Department of Medicine, Southern Arizona VA Health Care System, 3601 S 6Th Ave, Tucson, AZ 85723, USA.,Division of Hematology-Oncology, Department of Medicine, University of Arizona College of Medicine, 1501 North Campbell Avenue, Tucson, AZ 85724, USA.,Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, AZ 85724, USA
| | - Rebecca A Millius
- Department of Pathology, University of Arizona College of Medicine, 1501 North Campbell Avenue, Tucson, AZ 85724, USA
| | - Q Scott Ringenberg
- Division of Hematology-Oncology, Department of Medicine, Southern Arizona VA Health Care System, 3601 S 6Th Ave, Tucson, AZ 85723, USA
| | - Maria Bishop
- Division of Hematology-Oncology, Department of Medicine, Southern Arizona VA Health Care System, 3601 S 6Th Ave, Tucson, AZ 85723, USA.,Division of Hematology-Oncology, Department of Medicine, University of Arizona College of Medicine, 1501 North Campbell Avenue, Tucson, AZ 85724, USA.,Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, AZ 85724, USA
| |
Collapse
|
186
|
Polishchuk AL, Li R, Hill-Kayser C, Little A, Hawkins RA, Hamilton J, Lau M, Tran HC, Strahlendorf C, Lemons RS, Weinberg V, Matthay KK, DuBois SG, Marcus KJ, Bagatell R, Haas-Kogan DA. Likelihood of Bone Recurrence in Prior Sites of Metastasis in Patients With High-Risk Neuroblastoma. Int J Radiat Oncol Biol Phys 2014; 89:839-45. [DOI: 10.1016/j.ijrobp.2014.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 03/04/2014] [Accepted: 04/01/2014] [Indexed: 10/25/2022]
|
187
|
Gil TY, Lee DK, Lee JM, Yoo ES, Ryu KH. Clinical experience with (18)F-fluorodeoxyglucose positron emission tomography and (123)I-metaiodobenzylguanine scintigraphy in pediatric neuroblastoma: complementary roles in follow-up of patients. KOREAN JOURNAL OF PEDIATRICS 2014; 57:278-86. [PMID: 25076973 PMCID: PMC4115069 DOI: 10.3345/kjp.2014.57.6.278] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/16/2014] [Accepted: 05/16/2014] [Indexed: 12/04/2022]
Abstract
Purpose To evaluate the potential utility of 123I-metaiodobenzylguanine (123I-MIBG) scintigraphy and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) for the detection of primary and metastatic lesions in pediatric neuroblastoma (NBL) patients, and to determine whether 18F-FDG PET is as beneficial as 123I-MIBG imaging. Methods We selected 8 NBL patients with significant residual mass after operation and who had paired 123I-MIBG and 18F-FDG PET images that were obtained during the follow-up. We retrospectively reviewed the clinical charts and the findings of 45 paired scans. Results Both scans correlated relatively well with the disease status as determined by standard imaging modalities during follow-up; the overall concordance rates were 32/45 (71.1%) for primary tumor sites and 33/45 (73.3%) for bone-bone marrow (BM) metastatic sites. In detecting primary tumor sites, 123I-MIBG might be superior to 18F-FDG PET. The sensitivity of 123I-MIBG and 18F-FDG PET were 96.7% and 70.9%, respectively, and their specificity were 85.7% and 92.8%, respectively. 18F-FDG PET failed to detect 9 true NBL lesions in 45 follow-up scans (false negative rate, 29%) with positive 123I-MIBG. For bone-BM metastatic sites, the sensitivity of 123I-MIBG and 18F-FDG PET were 72.7% and 81.8%, respectively, and the specificity were 79.1% and 100%, respectively. 123I-MIBG scan showed higher false positivity (20.8%) than 18F-FDG PET (0%). Conclusion 123I-MIBG is superior for delineating primary tumor sites, and 18F-FDG PET could aid in discriminating inconclusive findings on bony metastatic NBL. Both scans can be complementarily used to clearly determine discrepancies or inconclusive findings on primary or bone-BM metastatic NBL during follow-up.
Collapse
Affiliation(s)
- Tae Young Gil
- Department of Pediatrics, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, Seoul, Korea
| | - Do Kyung Lee
- Department of Pediatrics, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, Seoul, Korea
| | - Jung Min Lee
- Department of Pediatrics, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, Seoul, Korea
| | - Eun Sun Yoo
- Department of Pediatrics, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, Seoul, Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, Seoul, Korea
| |
Collapse
|
188
|
Morgenstern DA, London WB, Stephens D, Volchenboum SL, Hero B, Di Cataldo A, Nakagawara A, Shimada H, Ambros PF, Matthay KK, Cohn SL, Pearson ADJ, Irwin MS. Metastatic neuroblastoma confined to distant lymph nodes (stage 4N) predicts outcome in patients with stage 4 disease: A study from the International Neuroblastoma Risk Group Database. J Clin Oncol 2014; 32:1228-35. [PMID: 24663047 DOI: 10.1200/jco.2013.53.6342] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE The presence of distant metastases is one of the most powerful predictors of outcome in patients with neuroblastoma. However, the pattern of metastatic spread is not incorporated into current risk stratification systems. Small case series have suggested that patients with neuroblastoma who have metastatic disease limited to distant lymph nodes (4N disease) may have improved outcomes. PATIENTS AND METHODS We analyzed retrospective data from the International Neuroblastoma Risk Group database for patients diagnosed from 1990 to 2002. 4N patients were compared with the remaining stage 4 patients (non-4N), excluding those with missing metastatic site data. RESULTS In all, 2,250 International Neuroblastoma Staging System stage 4 patients with complete data were identified, of whom 146 (6.5%) had 4N disease. For 4N patients, event-free survival (EFS; 5-year, 77% ± 4%) and overall survival (OS; 5-year, 85% ± 3%) were significantly better than EFS (5-year, 35% ± 1%) and OS (5-year, 42% ± 1%) for non-4N stage 4 patients (P < .001). 4N patients were more likely to be younger (P < .001) and have tumors with favorable characteristics, including absence of MYCN amplification (89% v 69%; P < .001). In a multivariable analysis, 4N disease remained a significant predictor of outcome (hazard ratio for non-4N v 4N: 3.40 for EFS and 3.69 for OS). Within subgroups defined by age at diagnosis and tumor MYCN status, 4N disease was significantly associated with improved outcomes. CONCLUSION 4N represents a subgroup with better outcome than that of other patients with metastatic disease. These findings suggest that the biology and treatment response of 4N tumors differ from other stage 4 tumors, and less intensive therapy should be considered for this cohort. Future exploration of biologic factors determining the pattern of metastatic spread is warranted.
Collapse
Affiliation(s)
- Daniel A Morgenstern
- Daniel A. Morgenstern and Meredith S. Irwin, Hospital for Sick Children and University of Toronto; Derek Stephens, Hospital for Sick Children, Toronto, ON, Canada; Daniel A. Morgenstern, Great Ormond Street Hospital for Children National Health Service (NHS) Foundation Trust, London; Andrew D.J. Pearson, The Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom; Wendy B. London, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA; Samuel L. Volchenboum and Susan L. Cohn, University of Chicago, Chicago, IL; Barbara Hero, University Children's Hospital, Köln, Germany; Andrea Di Cataldo, University of Catania, Catania, Italy; Akira Nakagawara, Chiba University School of Medicine, Chiba, Japan; Hiroyuki Shimada, University of Southern California at Los Angeles, Los Angeles; Katherine K. Matthay, University of California at San Francisco, San Francisco, CA; and Peter F. Ambros, St. Anna Kinderkrebsforschung, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Navarra M, Ursino MR, Ferlazzo N, Russo M, Schumacher U, Valentiner U. Effect of Citrus bergamia juice on human neuroblastoma cells in vitro and in metastatic xenograft models. Fitoterapia 2014; 95:83-92. [PMID: 24594241 DOI: 10.1016/j.fitote.2014.02.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 02/17/2014] [Accepted: 02/22/2014] [Indexed: 12/24/2022]
Abstract
Neuroblastoma is the most common extracranial pediatric solid tumor with poor prognosis in children with disseminated stage of disease. A number of studies show that molecules largely distributed in commonly consumed fruits and vegetables may have anti-tumor activity. In this study we evaluate the effect of Citrus bergamia (bergamot) juice (BJ) in vitro and in a spontaneous metastatic neuroblastoma SCID mouse model. Qualitative and quantitative characterizations of BJ flavonoid fractions were performed by RP-HPLC/PDA/MS. We show that BJ significantly affects SK-N-SH and LAN-1 cell proliferation in vitro, but fails to reduce primary tumor weight in vivo. Moreover, BJ reduced cell adhesiveness and invasion of LAN-1 and SK-N-SH cells in vitro and the number of pulmonary metastases under consideration of the number of tumor cells in the blood in mice inoculated with LAN-1 cells in vivo. These effects without any apparent sign of systemic toxicity confirm the potential clinical interest of BJ and lay the basis for further investigation in cancer.
Collapse
Affiliation(s)
- M Navarra
- Department of Drug Sciences and Health Products, University of Messina, Messina, Italy
| | - M R Ursino
- Department of Drug Sciences and Health Products, University of Messina, Messina, Italy
| | - N Ferlazzo
- Department of Drug Sciences and Health Products, University of Messina, Messina, Italy
| | - M Russo
- Department of Drug Sciences and Health Products, University of Messina, Messina, Italy
| | - U Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center, Hamburg-Eppendorf, Germany
| | - U Valentiner
- Institute of Anatomy and Experimental Morphology, University Medical Center, Hamburg-Eppendorf, Germany.
| |
Collapse
|
190
|
Schwankhaus N, Gathmann C, Wicklein D, Riecken K, Schumacher U, Valentiner U. Cell adhesion molecules in metastatic neuroblastoma models. Clin Exp Metastasis 2014; 31:483-96. [PMID: 24549749 DOI: 10.1007/s10585-014-9643-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 02/03/2014] [Indexed: 12/11/2022]
Abstract
Several cell adhesion molecules (CAMs) including selectins, integrins, cadherins and immunoglobulin-like CAMs are involved in leukocyte adhesion especially at sites of inflammation. In cancer cells, these CAMs have been associated with the growth and metastatic behavior in several malignant entities. In this study adhesion of LAN 1 and SK-N-SH neuroblastoma cells to selectins, hyaluronan and endothelial cells were determined under flow conditions. Furthermore cells were injected subcutaneously into wildtype and selectin deficient scid mice and their growth and metastatic behavior were analyzed. Under shear stress SK-N-SH cells firmly adhered to E-selectin-Fc-fusion protein, hyaluronan and endothelial cells, while LAN 1 cells showed less or hardly any adhesive events by comparison. In the SK-N-SH xenograft model metastasis formation was slightly dependent on the expression of selectins, while LAN 1 cells developed metastases completely independent of selectin expression. The different adhesive and metastatic properties of LAN 1 and SK-N-SH cells are reflected by a different expression profile of several CAMs. The results indicate that endothelial selectins are not essential for metastasis formation of human LAN 1 and SK-N-SH cells. However, other CAMs namely CD44, N-cadherin, NCAM and integrins were upregulated or downregulated, respectively, in SK-N-SH and LAN 1 cells and are potential adhesion molecules involved in the metastatic cascade of these cells.
Collapse
Affiliation(s)
- Nina Schwankhaus
- Center for Experimental Medicine, Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Christina Gathmann
- Center for Experimental Medicine, Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Daniel Wicklein
- Center for Experimental Medicine, Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Clinic for Stem Cell Transplantation, Center for Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Udo Schumacher
- Center for Experimental Medicine, Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ursula Valentiner
- Center for Experimental Medicine, Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
191
|
Affiliation(s)
- Daniel Y Wang
- Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA.
| | | |
Collapse
|
192
|
Kim PY, Tan O, Diakiw SM, Carter D, Sekerye EO, Wasinger VC, Liu T, Kavallaris M, Norris MD, Haber M, Chesler L, Dolnikov A, Trahair TN, Cheung NK, Marshall GM, Cheung BB. Identification of plasma complement C3 as a potential biomarker for neuroblastoma using a quantitative proteomic approach. J Proteomics 2013; 96:1-12. [PMID: 24200836 DOI: 10.1016/j.jprot.2013.10.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 10/16/2013] [Accepted: 10/22/2013] [Indexed: 11/26/2022]
Abstract
UNLABELLED The majority of patients diagnosed with neuroblastoma present with aggressive disease. Improved detection of neuroblastoma cancer cells following initial therapy may help in stratifying patient outcome and monitoring for relapse. To identify potential plasma biomarkers, we utilised a liquid chromatography-tandem mass spectrometry-based proteomics approach to detect differentially-expressed proteins in serum from TH-MYCN mice. TH-MYCN mice carry multiple copies of the human MYCN oncogene in the germline and homozygous mice for the transgene develop neuroblastoma in a manner resembling the human disease. The abundance of plasma proteins was measured over the course of disease initiation and progression. A list of 86 candidate plasma biomarkers was generated. Pathway analysis identified significant association of these proteins with genes involved in the complement system. One candidate, complement C3 protein, was significantly enriched in the plasma of TH-MYCN(+/+) mice at both 4 and 6weeks of age, and was found to be elevated in a cohort of human neuroblastoma plasma samples, compared to healthy subjects. In conclusion, we have demonstrated the suitability of the TH-MYCN(+/+) mouse model of neuroblastoma for identification of novel disease biomarkers in humans, and have identified Complement C3 as a candidate plasma biomarker for measuring disease state in neuroblastoma patients. BIOLOGICAL SIGNIFICANCE This study has utilised a unique murine model which develops neuroblastoma tumours that are biologically indistinguishable from human neuroblastoma. This animal model has effectively allowed the identification of plasma proteins which may serve as potential biomarkers of neuroblastoma. Furthermore, the label-free ion count quantitation technique which was used displays significant benefits as it is less labour intensive, feasible and accurate. We have been able to successfully validate this approach by confirming the differential abundance of two different plasma proteins. In addition, we have been able to confirm that the candidate biomarker Complement C3, is more abundant in the plasma of human neuroblastoma patient plasma samples when compared to healthy counterparts. Overall we have demonstrated that this approach can be potentially useful in the identification of biomarker candidates, and that further validation of the candidates may lead to the discovery of novel, clinically useful diagnostic tools in the detection of sub-clinical neuroblastoma.
Collapse
Affiliation(s)
- Patrick Y Kim
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | - Owen Tan
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | - Sonya M Diakiw
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | - Daniel Carter
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | - Eric O Sekerye
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | - Valerie C Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | - Maria Kavallaris
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | - Murray D Norris
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | - Lou Chesler
- Division of Cancer Biology, Institute for Cancer Research, Sutton, Surrey, UK
| | - Alla Dolnikov
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia
| | - Toby N Trahair
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia; Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW 2031, Australia
| | - Nai-Kong Cheung
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, United States
| | - Glenn M Marshall
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia; Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW 2031, Australia.
| | - Belamy B Cheung
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW 2031, Australia.
| |
Collapse
|
193
|
Maurer BJ, Kang MH, Janeba J, Villablanca JG, Groshen S, Matthay KK, Sondel PM, Maris JM, Jackson HA, Goodarzian F, Shimada H, Czarnecki S, Hasenauer B, Reynolds CP, Marachelian A. Phase I trial of fenretinide delivered orally in a novel organized lipid complex in patients with relapsed/refractory neuroblastoma: a report from the New Approaches to Neuroblastoma Therapy (NANT) consortium. Pediatr Blood Cancer 2013; 60:1801-8. [PMID: 23813912 PMCID: PMC4066886 DOI: 10.1002/pbc.24643] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 05/21/2013] [Indexed: 11/06/2022]
Abstract
BACKGROUND A phase I study was conducted to determine the maximum-tolerated dose, dose-limiting toxicities (DLTs), and pharmacokinetics of fenretinide (4-HPR) delivered in an oral powderized lipid complex (LXS) in patients with relapsed/refractory neuroblastoma. PROCEDURE 4-HPR/LXS powder (352-2,210 mg/m(2) /day) was administered on Days 0-6, in 21-day courses, by standard 3 + 3 design. RESULTS Thirty-two patients (median age = 8 years, range 3-27 years) enrolled with 30 evaluable for dose escalation. Prior therapies included stem cell transplantation/support (n = 26), 13-cis-retinoic acid (n = 22), (125/131) I-MIBG (n = 13), and anti-GD2 antibody (n = 6). 170+ courses were delivered. Course 1 DLTs were a Grade 3 (n = 1) alkaline phosphatase at 352 mg/m(2) /day. Other major toxicities were Grade 4 (n = 1) alkaline phosphatases on Courses 5 and 6 at 774 mg/m(2) /day, and Grade 3 (n = 1) ALT/AST elevation on Course 2 at 1,700 mg/m(2) /day. Of 29 response-evaluable patients, six had stable disease (SD) (4-26 courses); four with marrow- or bone disease-only had complete responses (CR) (10-46 courses). 4-HPR plasma levels were several folds higher (P < 0.05) than previously reported using capsular fenretinide. The Day 6 mean peak 4-HPR plasma level at 1,700 mg/m(2) /day was 21 µM. An MTD was not reached. CONCLUSIONS 4-HPR/LXS oral powder obtained higher plasma levels, with minimal toxicity and evidence of anti-tumor activity, than a previous capsule formulation. A recommended phase II schedule of 4-HPR/LXS powder is 1,500 mg/m(2) /day, TID, on Days 0-6, of a 21-day course.
Collapse
Affiliation(s)
- Barry J. Maurer
- Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, and Cancer Center, Lubbock TX,Department of Pediatrics, Texas Tech University Health Sciences Center, and Cancer Center, Lubbock TX,Department of Medicine, Texas Tech University Health Sciences Center, and Cancer Center, Lubbock TX
| | - Min H. Kang
- Department of Pharmacology, Texas Tech University Health Sciences Center, and Cancer Center, Lubbock TX,Department of Medicine, Texas Tech University Health Sciences Center, and Cancer Center, Lubbock TX
| | - Jitka Janeba
- Saban Research Institute, University of Southern California, Los Angeles, CA
| | | | - Susan Groshen
- Department of Statistics, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Katherine K. Matthay
- Department of Pediatrics, University of California San Francisco School of Medicine, San Francisco, CA
| | - Paul M. Sondel
- University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, Madison, WI
| | - John M. Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, University of Pennsylvania and School of Medicine and Abramson Family Cancer Research Institute, Philadelphia, PA
| | - Hollie A. Jackson
- Department of Radiology, University of Southern California, Los Angeles, CA
| | - Fariba Goodarzian
- Department of Radiology, University of Southern California, Los Angeles, CA
| | - Hiroyuki Shimada
- Department of Pathology, University of Southern California, Los Angeles, CA
| | - Scarlett Czarnecki
- New Approaches to Neuroblastoma Therapy (NANT) Consortium Operations Center, Childrens Hospital Los Angeles, Los Angeles, CA
| | - Beth Hasenauer
- New Approaches to Neuroblastoma Therapy (NANT) Consortium Operations Center, Childrens Hospital Los Angeles, Los Angeles, CA
| | - C. Patrick Reynolds
- Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, and Cancer Center, Lubbock TX,Department of Pediatrics, Texas Tech University Health Sciences Center, and Cancer Center, Lubbock TX,Department of Medicine, Texas Tech University Health Sciences Center, and Cancer Center, Lubbock TX
| | - Araz Marachelian
- Department of Pediatrics, University of Southern California, Los Angeles, CA
| |
Collapse
|
194
|
Jamil S, Hultman I, Cedervall J, Ali RQ, Fuchs G, Gustavsson B, Asmundsson J, Sandstedt B, Kogner P, Ährlund-Richter L. Tropism of thein situgrowth from biopsies of childhood neuroectodermal tumors following transplantation into experimental teratoma. Int J Cancer 2013; 134:1630-7. [DOI: 10.1002/ijc.28498] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/02/2013] [Indexed: 11/07/2022]
Affiliation(s)
- Seema Jamil
- Department of Women's and Children's Health; Karolinska Institutet; Stockholm Sweden
| | - Isabell Hultman
- Department of Women's and Children's Health; Karolinska Institutet; Stockholm Sweden
| | - Jessica Cedervall
- Department of Medical Biochemistry and Microbiology; Uppsala University; Uppsala Sweden
| | | | - Gabriel Fuchs
- Department of Women's and Children's Health; Karolinska Institutet; Stockholm Sweden
| | - Bengt Gustavsson
- Department of Oncology and Pathology; Karolinska Institutet; Stockholm Sweden
| | - Jurate Asmundsson
- Department of Oncology and Pathology; Karolinska Institutet; Stockholm Sweden
- Karolinska University Laboratories, Unit for Pathology; Stockholm Sweden
| | - Bengt Sandstedt
- Department of Women's and Children's Health; Karolinska Institutet; Stockholm Sweden
- Karolinska University Laboratories, Unit for Pathology; Stockholm Sweden
| | - Per Kogner
- Department of Women's and Children's Health; Karolinska Institutet; Stockholm Sweden
| | - Lars Ährlund-Richter
- Department of Women's and Children's Health; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
195
|
Lee HJ, Park MK, Bae HC, Yoon HJ, Kim SY, Lee CH. Transglutaminase-2 Is Involved in All-Trans Retinoic Acid-Induced Invasion and Matrix Metalloproteinases Expression of SH-SY5Y Neuroblastoma Cells via NF-κB Pathway. Biomol Ther (Seoul) 2013; 20:286-92. [PMID: 24130925 PMCID: PMC3794525 DOI: 10.4062/biomolther.2012.20.3.286] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 04/30/2012] [Accepted: 05/03/2012] [Indexed: 11/28/2022] Open
Abstract
All-trans retinoic acid (ATRA) is currently used in adjuvant differentiation-based treatment of residual or relapsed neuroblastoma (NB). It has been reported that short-term ATRA treatment induces migration and invasion of SH-SY5Y via transglutaminase-2 (Tgase-2). However, the detailed mechanism of Tgase-2's involvement in NB cell invasion remains unclear. Therefore we investigated the role of Tgase-2 in invasion of NB cells using SH-SY5Y cells. ATRA dose-dependently induced the invasion of SH-SY5Y cells. Cystamine (CTM), a well known tgase inhibitor suppressed the ATRA-induced invasion of SH-SY5Y cells in a dose-dependent manner. Matrix metalloproteinase-9 (MMP-9) and MMP-2, well known genes involved in invasion of cancer cells were induced in the ATRA-induced invasion of the SH-SH5Y cells. Treatment of CTM suppressed the MMP-9 and MMP-2 enzyme activities in the ATRA-induced invasion of the SH-SY5Y cells. To confirm the involvement of Tgase-2, gene silencing of Tgase-2 was performed in the ATRA-induced invasion of the SH-SH5Y cells. The siRNA of Tgase-2 suppressed the MMP-9 and MMP-2 activity of the SH-SY5Y cells. MMP-2 and MMP-9 are well known target genes of NF-κB. Therefore the relationship of Tgase-2 and NF-κB in the ATRA-induced invasion of the SH-SY5Y cells was examined using siRNA and CTM. ATRA induced the activation of NF-κB in the SH-SY5Y cells and CTM suppressed the activation of NF-κB. Gene silencing of Tgase-2 suppressed the MMP expression by ATRA. These results suggested that Tgase-2 might be a new target for controlling the ATRA-induced invasion of NBs.
Collapse
Affiliation(s)
- Hye Ja Lee
- College of Pharmacy, Dongguk University, Seoul 100-715
| | | | | | | | | | | |
Collapse
|
196
|
Abstract
Neuroblastoma, the most common extracranial solid tumor of childhood, is thought to originate from undifferentiated neural crest cells. Amplification of the MYC family member, MYCN, is found in ∼25% of cases and correlates with high-risk disease and poor prognosis. Currently, amplification of MYCN remains the best-characterized genetic marker of risk in neuroblastoma. This article reviews roles for MYCN in neuroblastoma and highlights recent identification of other driver mutations. Strategies to target MYCN at the level of protein stability and transcription are also reviewed.
Collapse
Affiliation(s)
- Miller Huang
- Departments of Neurology, Pediatrics, and Neurosurgery, University of California, San Francisco, California 94158-9001
| | | |
Collapse
|
197
|
Kose D, Karabagli P, Yavas G, Karabagli H, Koksal Y. Intracranial metastasis of neuroblastoma: in two different areas at the same time. Childs Nerv Syst 2013; 29:1799-802. [PMID: 23793924 DOI: 10.1007/s00381-013-2203-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 06/05/2013] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Neuroblastoma (NB) is the most common extracranial solid malignancy in children. The major cause of death from this cancer is metastasis of tumors, and metastasis can be seen in different areas of the body. Metastasis of NB occurs by hematogenous and lymphatic routes. Generally, brain metastases have been reported in only one area of the brain parenchyma. CASE REPORT A 4-year-old male patient was treated in our clinic due to the NB that settled in the intra-abdominal region, but the patient presented with headache and nausea approximately 5 months after completion of the treatment. Whereupon, cranial imaging was performed and two masses were detected in the bilateral frontal lobe, and then the patient underwent surgery. Metastatic NB diagnosis was confirmed histopathologically. The patient's chemotherapy treatment is still ongoing. CONCLUSION We report the case of a male patient with two separate metastatic masses in the brain parenchyma, which occurred in two different areas at the same time.
Collapse
Affiliation(s)
- Dogan Kose
- Faculty of Medicine, Department of Pediatric Hematology and Oncology, Selcuk University, Konya, Turkey,
| | | | | | | | | |
Collapse
|
198
|
Ara T, Nakata R, Sheard MA, Shimada H, Buettner R, Groshen SG, Ji L, Yu H, Jove R, Seeger RC, DeClerck YA. Critical role of STAT3 in IL-6-mediated drug resistance in human neuroblastoma. Cancer Res 2013. [PMID: 23633489 DOI: 10.1158/0008-5472.can-12-2353.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Drug resistance is a major cause of treatment failure in cancer. Here, we have evaluated the role of STAT3 in environment-mediated drug resistance (EMDR) in human neuroblastoma. We determined that STAT3 was not constitutively active in most neuroblastoma cell lines but was rapidly activated upon treatment with interleukin (IL)-6 alone and in combination with the soluble IL-6 receptor (sIL-6R). Treatment of neuroblastoma cells with IL-6 protected them from drug-induced apoptosis in a STAT3-dependent manner because the protective effect of IL-6 was abrogated in the presence of a STAT3 inhibitor and upon STAT3 knockdown. STAT3 was necessary for the upregulation of several survival factors such as survivin (BIRC5) and Bcl-xL (BCL2L1) when cells were exposed to IL-6. Importantly, IL-6-mediated STAT3 activation was enhanced by sIL-6R produced by human monocytes, pointing to an important function of monocytes in promoting IL-6-mediated EMDR. Our data also point to the presence of reciprocal activation of STAT3 between tumor cells and bone marrow stromal cells including not only monocytes but also regulatory T cells (Treg) and nonmyeloid stromal cells. Thus, the data identify an IL-6/sIL-6R/STAT3 interactive pathway between neuroblastoma cells and their microenvironment that contributes to drug resistance.
Collapse
Affiliation(s)
- Tasnim Ara
- Division of Hematology-Oncology, Department of Pediatrics, Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Roles of full-length and truncated neurokinin-1 receptors on tumor progression and distant metastasis in human breast cancer. Breast Cancer Res Treat 2013; 140:49-61. [DOI: 10.1007/s10549-013-2599-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 06/06/2013] [Indexed: 12/15/2022]
|
200
|
Sridhar S, Bhadada SK, Bhansali A, Bhattacharya A. Lytic and sclerotic (mixed) vertebral metastasis in ganglioneuroblastoma. Indian J Nucl Med 2013; 27:127-9. [PMID: 23723589 PMCID: PMC3665142 DOI: 10.4103/0972-3919.110721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The clinical presentation of ganglioneuroblastoma is highly variable and it is not uncommon to see metastasis at presentation. Bone is the second most common site of metastasis in neuroblastoma. Neuroblastoma cells usually activate osteoclasts and form osteolytic lesions. Here, we describe a patient who presented with back pain. On evaluation, X-ray and positron emission tomography-computed tomography showed mixed lytic and sclerotic vertebral metastasis, and subsequently diagnosed as ganglioneuroblastoma.
Collapse
|