Skip to main content
Top
Published in: Clinical & Experimental Metastasis 3/2011

01-03-2011 | Research Paper

Expression and significance of HER family receptors in neuroblastic tumors

Authors: Ewa Izycka-Swieszewska, Agnieszka Wozniak, Elzbieta Drozynska, Jacek Kot, Wieslawa Grajkowska, Teresa Klepacka, Danuta Perek, Sylwia Koltan, Ewa Bien, Janusz Limon

Published in: Clinical & Experimental Metastasis | Issue 3/2011

Login to get access

Abstract

HER receptor family plays an important role in normal embryonic development and is involved in pathogenesis and progression of some types of cancer. Neuroblastic tumors (NT) are common pediatric neoplasms with a poor outcome in a significant number of patients. The biological and prognostic role of HER family in NT is not well established. In the current study we evaluated HER1–4 receptors expression, their prognostic significance and clinicopathological correlations in a series of 103 NTs by immunohistochemical assessment of HER1–4 expression and FISH analysis of EGFR and HER2 copy number status. HER receptors are commonly expressed in NT but it was not due to EGFR or HER2 amplification. EGFR, HER2 and HER4 show correlation with tumor histology. It seems that these receptors take part in neuroblastic cell differentiation and Schwannian stroma development. EGFR and HER2 positivity are more frequently found in favorable histological risk group of tumours (P = 0.004 and P = 0.01 respectively) while high expression of HER4 is significantly more often found in patients with metastatic disease (P = 0.03). Moreover tumors with HER2 polysomy were more often found in children ≤18 months, with localized disease, and favorable histological group. Our study showed that the role of HER family members in NT biology is interrelated and complex but their expression level may present a novel prognostic factor for NT patients outcome.
Literature
1.
go back to reference Brodeur G (2003) Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer 3:203–216CrossRefPubMed Brodeur G (2003) Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer 3:203–216CrossRefPubMed
3.
go back to reference Nakagawara A (2004) Neural crest development and neuroblastoma: the genetic and biological link. Prog Brain Res 146:233–242PubMed Nakagawara A (2004) Neural crest development and neuroblastoma: the genetic and biological link. Prog Brain Res 146:233–242PubMed
4.
go back to reference Mosse YP, Laudenslager M, Longo L, Maris J (2008) Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 455:930–935CrossRefPubMed Mosse YP, Laudenslager M, Longo L, Maris J (2008) Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 455:930–935CrossRefPubMed
5.
go back to reference Shimada H, Ambros I, Dehner L et al (1999) The international neuroblastoma pathology classification (the Shimada system). Cancer 86:364–372CrossRefPubMed Shimada H, Ambros I, Dehner L et al (1999) The international neuroblastoma pathology classification (the Shimada system). Cancer 86:364–372CrossRefPubMed
6.
go back to reference Joshi W, Silverman J, Altshuler G et al (1993) Systematization of primary histopathologic and fine-needle aspiration cytologic features and description of unusual histopathologic feature of neuroblastic tumors: a report from the Pediatric Oncology Group. Hum Pathol 24:493–504CrossRefPubMed Joshi W, Silverman J, Altshuler G et al (1993) Systematization of primary histopathologic and fine-needle aspiration cytologic features and description of unusual histopathologic feature of neuroblastic tumors: a report from the Pediatric Oncology Group. Hum Pathol 24:493–504CrossRefPubMed
7.
go back to reference Cohn S, Pearson A, London W et al (2009) The international neuroblastoma risk group (INRG) classification system: an INRG task force report. J Clin Oncol 27:289–297CrossRefPubMed Cohn S, Pearson A, London W et al (2009) The international neuroblastoma risk group (INRG) classification system: an INRG task force report. J Clin Oncol 27:289–297CrossRefPubMed
8.
go back to reference Fong A, Park J (2009) High-risk neuroblastoma: a therapy in evolution. Pediatr Hematol Oncol 26:539–548CrossRefPubMed Fong A, Park J (2009) High-risk neuroblastoma: a therapy in evolution. Pediatr Hematol Oncol 26:539–548CrossRefPubMed
9.
go back to reference Jakacki R, Hamilton M, Gilbertson R et al (2008) Pediatric phase I and pharmacokinetic study of Erlotinib followed by the combination of Erlotinib and Temozolomid: a children’s oncology group phase I consortium study. J Clin Oncol 26:4921–4927CrossRefPubMed Jakacki R, Hamilton M, Gilbertson R et al (2008) Pediatric phase I and pharmacokinetic study of Erlotinib followed by the combination of Erlotinib and Temozolomid: a children’s oncology group phase I consortium study. J Clin Oncol 26:4921–4927CrossRefPubMed
10.
go back to reference Vermuelen J, De Preter K, Naranjo A et al (2009) Predicting outcomes for children with neuroblastoma using a multigene expression signature: a retrospective SIOPEN/COG/GPOH study. Lancet Oncol 10:663–671CrossRef Vermuelen J, De Preter K, Naranjo A et al (2009) Predicting outcomes for children with neuroblastoma using a multigene expression signature: a retrospective SIOPEN/COG/GPOH study. Lancet Oncol 10:663–671CrossRef
11.
go back to reference Baselga J, Swain S (2009) Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer 9:463–475CrossRefPubMed Baselga J, Swain S (2009) Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer 9:463–475CrossRefPubMed
12.
go back to reference Citri A, Yarden Y (2006) EGF-ERBB signaling: towards the systems level. Nat Rev Mol Cell Biol 7:505–516CrossRefPubMed Citri A, Yarden Y (2006) EGF-ERBB signaling: towards the systems level. Nat Rev Mol Cell Biol 7:505–516CrossRefPubMed
13.
go back to reference Zaczek A, Brandt B, Bielawski KP (2005) The diverse signaling network of EGFR, HER2, HER3 and HER4 tyrosine kinase receptors and the consequences for therapeutic approaches. Histol Histopathol 20:1005–1015PubMed Zaczek A, Brandt B, Bielawski KP (2005) The diverse signaling network of EGFR, HER2, HER3 and HER4 tyrosine kinase receptors and the consequences for therapeutic approaches. Histol Histopathol 20:1005–1015PubMed
14.
go back to reference Britsch S, Li L, Kirchhoff S et al (1998) The ErbB2 and ErbB3 receptors and their ligand, neuregulin-1, are essential for development of the sympathetic nervous system. Genes Dev 12:1825–1836CrossRefPubMed Britsch S, Li L, Kirchhoff S et al (1998) The ErbB2 and ErbB3 receptors and their ligand, neuregulin-1, are essential for development of the sympathetic nervous system. Genes Dev 12:1825–1836CrossRefPubMed
15.
go back to reference Casalini P, Iorio M, Galmozzi E, Menard S (2004) Role of HER receptors family in development and differentiation. J Cell Physiol 200:343–350CrossRefPubMed Casalini P, Iorio M, Galmozzi E, Menard S (2004) Role of HER receptors family in development and differentiation. J Cell Physiol 200:343–350CrossRefPubMed
16.
go back to reference Chan R, Hardy W, Laing M, Hardy S, Muller W (2002) The catalytic activity of the ErbB2 receptor tyrosine kinase is essential for embryonic development. Mol Cell Biol 22:1073–1078CrossRefPubMed Chan R, Hardy W, Laing M, Hardy S, Muller W (2002) The catalytic activity of the ErbB2 receptor tyrosine kinase is essential for embryonic development. Mol Cell Biol 22:1073–1078CrossRefPubMed
17.
go back to reference Ganti R, Skapek S, Zhang J et al (2006) Expression and genomic status of EGFR and ErbB-2 in alveolar and embryonal rhabdomyosarcoma. Modern Pathol 19:1213–1220CrossRef Ganti R, Skapek S, Zhang J et al (2006) Expression and genomic status of EGFR and ErbB-2 in alveolar and embryonal rhabdomyosarcoma. Modern Pathol 19:1213–1220CrossRef
18.
go back to reference Ho R, Minturn J, Hishiki T et al (2005) Proliferation of human neuroblastomas mediated by the epidermal growth factor receptor. Cancer Res 65:9868–9875CrossRefPubMed Ho R, Minturn J, Hishiki T et al (2005) Proliferation of human neuroblastomas mediated by the epidermal growth factor receptor. Cancer Res 65:9868–9875CrossRefPubMed
19.
go back to reference Richards KN, Zweidler-McKay PA, Van Roy N et al (2010) Signaling of ERBB receptor tyrosine kinases promotes neuroblastoma growth in vitro and in vivo. Cancer 116:3233–3243CrossRefPubMed Richards KN, Zweidler-McKay PA, Van Roy N et al (2010) Signaling of ERBB receptor tyrosine kinases promotes neuroblastoma growth in vitro and in vivo. Cancer 116:3233–3243CrossRefPubMed
20.
go back to reference Qualman S, Bowen J, Fitzgibbons P et al (2005) Protocol for the examination of specimens from patients with neuroblastoma and related neuroblastic tumors. Arch Pathol Lab Med 129:874–883PubMed Qualman S, Bowen J, Fitzgibbons P et al (2005) Protocol for the examination of specimens from patients with neuroblastoma and related neuroblastic tumors. Arch Pathol Lab Med 129:874–883PubMed
21.
go back to reference Izycka-Swieszewska E, Wozniak A, Kot J et al (2010) Prognostic significance of HER2 expression in neuroblastic tumors. Mod Pathol 23:1261–1268CrossRefPubMed Izycka-Swieszewska E, Wozniak A, Kot J et al (2010) Prognostic significance of HER2 expression in neuroblastic tumors. Mod Pathol 23:1261–1268CrossRefPubMed
22.
go back to reference Corzo C, Bellosillo B, Corominas J et al (2007) Does polysomy of chromosome 17 have a role in ERBB2 and topoisomerase IIα expression. Tumor Biol 28:221–228CrossRef Corzo C, Bellosillo B, Corominas J et al (2007) Does polysomy of chromosome 17 have a role in ERBB2 and topoisomerase IIα expression. Tumor Biol 28:221–228CrossRef
23.
go back to reference Varella-Garcia M (2006) Stratification of non-small cell lung cancer patients for therapy with epidermal growth factor receptor inhibitors: the EGFR fluorescence in situ hybridization assay. Diagn Pathol 1:19CrossRefPubMed Varella-Garcia M (2006) Stratification of non-small cell lung cancer patients for therapy with epidermal growth factor receptor inhibitors: the EGFR fluorescence in situ hybridization assay. Diagn Pathol 1:19CrossRefPubMed
24.
go back to reference Vandensompele J, Baudis M, De Preter K et al (2005) Unequivocal delineation of clinicogenetic subgroups and development of a new model for improved outcome prediction in neuroblastoma. J Clin Oncol 23:2280–2299CrossRef Vandensompele J, Baudis M, De Preter K et al (2005) Unequivocal delineation of clinicogenetic subgroups and development of a new model for improved outcome prediction in neuroblastoma. J Clin Oncol 23:2280–2299CrossRef
25.
go back to reference Gilbertson RJ (2005) ERBB2 in pediatric cancer: innocent until proven guilty. Oncologist 10:508–517CrossRefPubMed Gilbertson RJ (2005) ERBB2 in pediatric cancer: innocent until proven guilty. Oncologist 10:508–517CrossRefPubMed
26.
go back to reference Rogers S, Harrington K, Rhys-Evans P et al (2005) Biological significance of c-erbB family oncogenes in head and neck cancer. Cancer Metastasis Rev 24:47–69CrossRefPubMed Rogers S, Harrington K, Rhys-Evans P et al (2005) Biological significance of c-erbB family oncogenes in head and neck cancer. Cancer Metastasis Rev 24:47–69CrossRefPubMed
27.
go back to reference Layfield L, Thompson K, Dodge R, Kerns B (1995) Prognostic indicators for neuroblastoma: stage, grade, DNA ploidy, MIB-1-proliferation index, p53, HER-2/neu and EGFR—a survival study. J Surg Oncol 59:21–27CrossRefPubMed Layfield L, Thompson K, Dodge R, Kerns B (1995) Prognostic indicators for neuroblastoma: stage, grade, DNA ploidy, MIB-1-proliferation index, p53, HER-2/neu and EGFR—a survival study. J Surg Oncol 59:21–27CrossRefPubMed
28.
go back to reference Tamura S, Hosoi H, Kuwahara Y et al (2007) Induction of apoptosis by an inhibitor of EGFR in neuroblastoma cells. BBRC 358:226–232PubMed Tamura S, Hosoi H, Kuwahara Y et al (2007) Induction of apoptosis by an inhibitor of EGFR in neuroblastoma cells. BBRC 358:226–232PubMed
29.
go back to reference Rossler J, Odenthal E, Geoerger B et al (2009) EGFR inhibition using Gefitinib is not active in neuroblastoma cell lines. Anticancer Res 29:1327–1334PubMed Rossler J, Odenthal E, Geoerger B et al (2009) EGFR inhibition using Gefitinib is not active in neuroblastoma cell lines. Anticancer Res 29:1327–1334PubMed
30.
go back to reference Chiu B, Mirkin B, Madonna M (2007) Epidermal growth factor can induce apoptosis in neuroblastoma. Mitogenic and apoptotic actions of EGF on neuroblastoma cells are concentration-dependent. J Ped Surg 42:482–488CrossRef Chiu B, Mirkin B, Madonna M (2007) Epidermal growth factor can induce apoptosis in neuroblastoma. Mitogenic and apoptotic actions of EGF on neuroblastoma cells are concentration-dependent. J Ped Surg 42:482–488CrossRef
31.
go back to reference Tovey S, Reeves J, Stanton P, Ozanne B, Barlett J, Cooke T (2006) Low expression of HER2 protein in breast cancer is biologically significant. J Pathol 210:358–362CrossRefPubMed Tovey S, Reeves J, Stanton P, Ozanne B, Barlett J, Cooke T (2006) Low expression of HER2 protein in breast cancer is biologically significant. J Pathol 210:358–362CrossRefPubMed
32.
go back to reference Goji J, Nakamura H, Ito H, Mabuchi O, Hashimoto K, Sano K (1995) Expression of c-ErbB2 in human neuroblastoma tissues, adrenal medulla adjacent to tumor, and developing mouse neural crest cells. Am J Pathol 146:660–672PubMed Goji J, Nakamura H, Ito H, Mabuchi O, Hashimoto K, Sano K (1995) Expression of c-ErbB2 in human neuroblastoma tissues, adrenal medulla adjacent to tumor, and developing mouse neural crest cells. Am J Pathol 146:660–672PubMed
33.
go back to reference Gambini C, Sementa AR, Boni L et al (2003) Expression of HER2/neu is uncommon in human neuroblastic tumors and is unrelated to tumor progression. Cancer Immunol Immunother 52:116–120PubMed Gambini C, Sementa AR, Boni L et al (2003) Expression of HER2/neu is uncommon in human neuroblastic tumors and is unrelated to tumor progression. Cancer Immunol Immunother 52:116–120PubMed
34.
go back to reference Hayashi M, Inokuchi M, Takagi Y et al (2008) High expression of HER3 is associated with a decreased survival in gastric cancer. Clin Cancer Res 14:7843–7849CrossRefPubMed Hayashi M, Inokuchi M, Takagi Y et al (2008) High expression of HER3 is associated with a decreased survival in gastric cancer. Clin Cancer Res 14:7843–7849CrossRefPubMed
35.
go back to reference Witton C, Reeves J, Going J, Cooke T, Barlett J (2003) Expression of the HER1–4 family of receptor tyrosine kinases in breast cancer. J Pathol 200:290–297CrossRefPubMed Witton C, Reeves J, Going J, Cooke T, Barlett J (2003) Expression of the HER1–4 family of receptor tyrosine kinases in breast cancer. J Pathol 200:290–297CrossRefPubMed
36.
go back to reference Srinivasan R, Poulsom R, Hurst H, Gullick W (1998) Expression of the c-erbB-4/HER4 protein and mRNA in normal human fetal and adult tissues and in a survey of nine solid tumor types. J Pathol 185:236–245CrossRefPubMed Srinivasan R, Poulsom R, Hurst H, Gullick W (1998) Expression of the c-erbB-4/HER4 protein and mRNA in normal human fetal and adult tissues and in a survey of nine solid tumor types. J Pathol 185:236–245CrossRefPubMed
37.
go back to reference Muraoka-Cook R, Sandahl M, Husted C et al (2006) The intracellular domain of ErbB4 induces differentiation of mammary epithelial cells. Mol Biol Cell 17:4118–4129CrossRefPubMed Muraoka-Cook R, Sandahl M, Husted C et al (2006) The intracellular domain of ErbB4 induces differentiation of mammary epithelial cells. Mol Biol Cell 17:4118–4129CrossRefPubMed
Metadata
Title
Expression and significance of HER family receptors in neuroblastic tumors
Authors
Ewa Izycka-Swieszewska
Agnieszka Wozniak
Elzbieta Drozynska
Jacek Kot
Wieslawa Grajkowska
Teresa Klepacka
Danuta Perek
Sylwia Koltan
Ewa Bien
Janusz Limon
Publication date
01-03-2011
Publisher
Springer Netherlands
Published in
Clinical & Experimental Metastasis / Issue 3/2011
Print ISSN: 0262-0898
Electronic ISSN: 1573-7276
DOI
https://doi.org/10.1007/s10585-010-9369-1

Other articles of this Issue 3/2011

Clinical & Experimental Metastasis 3/2011 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine