Purpose: Von Hippel-Lindau (VHL) disease is an inherited syndrome caused by germline mutations in the VHL tumor suppressor gene, predisposing to a variety of neoplasms including pancreatic neuroendocrine tumors (PanNET). In VHL disease, PanNET probably progress according to a specific pathway of carcinogenesis. Our aim was to characterize by molecular quantitative analysis a panel of molecules implicated in the VHL pathway and in tumor progression in the PanNET of patients with VHL.

Experimental Design: The expression of 52 genes was studied by quantitative reverse transcriptase PCR in 18 patients with VHL operated on for PanNET and compared with 16 non-VHL PanNET. The VHL and non-VHL tumors were matched according to their size and cell proliferation. For some genes, we looked for differences in the protein expression in VHL PanNET (n = 31), microadenomas (n = 22), and non-VHL PanNET (n = 16), included in tissue microarray blocks.

Results: Nineteen (36%) genes were significantly upregulated and three (6%) downregulated in VHL PanNET. The upregulated genes were related to (i) hypoxia-inducible factor (HIF) molecules (CA9, HIF2A, and GLUT1), (ii) angiogenesis (CDH5, VEGFR1, EDNRA, ANGPT2, CD34, VEGFR2, VEGFA, and ANGPT1), (iii) the processes of epithelial–mesenchymal transition (VIM) and/or metastasis (LAMA4 and CXCR4), (iv) growth factors and receptors (PDGFB, IRS1, and ERBB1), or (v) cell cycle (CCND1 and CDKN2A). The downregulated genes were related to (i) EMT (OCLN) and (ii) signaling pathways (RPS6KB1 and GADD45B).

Conclusion: This study shows that the progression of PanNET in patients with VHL tumors follows a specific pathway and supports that targeting molecules specifically involved may be of therapeutic importance. Clin Cancer Res; 18(10); 2838–49. ©2012 AACR.

Translational Relevance

Patients with VHL disease can present pancreatic neuroendocrine tumors (PanNET), which can be associated with microadenomas. VHL PanNET are very angiogenic, frequently locally aggressive, and present different characteristics as compared with sporadic tumors.

By molecular quantitative analysis, we found a panel of genes differentially regulated between VHL and non-VHL PanNET. Some of these genes are involved in angiogenesis, epithelial–mesenchymal transition, progression of metastasis, and cell signalling; most of them may have important biologic and clinical relevance.

Our study might impact on clinical practice as it suggests that the development of VHL PanNET follows a specific pathway and supports that targeting molecules specifically involved (e.g., with antiangiogenic or CXCR4 chemokine antagonist…) may be of therapeutic importance.

Von Hippel-Lindau (VHL) disease is an autosomal dominant disorder caused by a germline mutation in the VHL tumor suppressor gene, located in chromosome 3p25. Affected individuals are predisposed to develop a variety of neoplasms characterized by their marked phenotypic variability and the age-dependent penetrance (1, 2). Pancreatic neuroendocrine tumors (PanNET) develop in 10% to 17% of patients with VHL, and present specifics and differences compared with sporadic PanNET, including multiplicity, association with microadenomas and a high frequency of local tumor aggressiveness at histologic and macroscopic levels, showing peripancreatic organ invasion in 20% of the patients (1–4).

The VHL gene encodes a protein (pVHL) involved in the regulation of a transcription factor called hypoxia-inducible factor (HIF). Mutations within the VHL gene disrupt the interaction between HIF-α and pVHL, leading to constitutive HIF activation and expression of HIF targets, involved in many cellular processes, including angiogenesis and cell metabolism (1, 2). VHL also has independent effects of its role in degradation of HIF-α subunits.

In VHL disease, PanNET probably develop according to a specific pathway of carcinogenesis. To obtain further insight into this process, we used real-time quantitative reverse transcriptase PCR (RT-PCR) to quantify the mRNA expression of a large number of selected genes in VHL tumors in comparison with sporadic tumors. We particularly focused on the expression of genes related to angiogenesis, to the HIF/VHL pathway, to the processes of epithelial–mesenchymal transition (EMT) or metastasis, and involved in cellular mechanisms such as cell cycle and signaling (2, 5–8). Genes of interest were further studied at the protein level by immunohistochemistry and compared with microadenomas, which represent the early neuroendocrine tumor precursors.

Patients and tumors

Thirty-one patients with VHL (19 women and 12 men) that underwent pancreatic surgery for one or several PanNET between April 1982 and February 2009 were retrospectively studied (Table 1). In comparison with the previous publication of our group which described the pathologic data of the pancreas of 18 patients with VHL treated in Beaujon Hospital (Clichy, France), we added 13 cases in this study, 1 from Beaujon hospital (Clichy) and 12 from different cities of France (2). The median (range) age of patients at the time of surgery was 40.7 years (25–76). Surgery consisted of pancreaticoduodenectomy in 19 patients, distal pancreatectomy in 9 patients, 1 total pancreatectomy, and 2 limited resections of the pancreas.

Table 1.

Clinical and histopathologic data for the VHL and non-VHL patients

PatientSex/ageSurgical procedureNumber of mAdTumors number/size, cmWHO grade (% Ki-67)TNMFollow-up, moStatusVHL diseaseFrozen tissue/PCR
F/37 PD 1/5 G1 (1) pT3N1M0 155 AFD 
F/28 PD >15 3/0.7–1.5 G1 (1) pT1mN0M0 170 AFD 
F/34 PD 2/1.2–3.2 G1 (1) pT2mN1M0 116 DOD 
M/39 LP 1/3 G1 (1) pT2N0M0 112 DOC 
M/46 PD >15 4/1–3 G1 (2) pT2mN1M0 125 AWD 
F/49 LP 1/2 G2 (10) pT2N0M0 DOC 
M/26 PD >15 2/2 G1 (1) pT2mN0M0 72 AFD 
F/26 PD >15 3/0.6–2.5 G2 (6) pT2mN0M0 44 AFD 
M/51 PD 3/0.8–4.5 G2 (5) pT3mN0M0 13 DOC 
10 F/33 PD 1/2 G2 (5) pT2N1M0 115 AFD 
11 F/28 LP 1/3 G2 (8) pT2N0M0 97 AFD 
12 M/43 LP >15 4/1–4 G2 (7) pT2mN0M0 96 AFD 
13 F/56 LP 1/3.4 G2 (7) pT2N1M0 99 AFD 
14 M/48 PD 5/0.7–2.5 G2 (8) pT2mN1M0 47 AFD 
15 M/30 PD 1/2.5 G2 (5) pT2N0M0 48 AFD 
16 F/33 PD 2/1.3–3.4 G2 (12) pT2mN0M0 60 AFD 
17 F/50 PD 1/5 G1 (1) pT3N1M0 66 AWD 
18 F/35 PD 1/3.2 G2 (5) pT2N0M0 38 AFD 
19 M/26 PD 2/0.6–2.5 G1 (2) pT2mN1M0 87 AWD − 
20 F/44 PD 1/4.2 G2 (8) pT3N0M0 79 AWD − 
21 F/48 PD 3/0.6–2.5 G2 (10) pT2mN0M0 89 AWD − 
22 F/25 PD 3/0.6–2 G1 (1) pT2mN0M0 137 AFD − 
23 F/25 LP 2/0.6–0.8 G1 (1) pT1mN0M1 168 DOC − 
24 F/48 LP 1/4 G1 (1) pT2N0M0 65 AFD − 
25 M/34 EN 1/2 G2 (3) pT2N0M0 12 AFD − 
26 F/76 TP >10 3/1–4 G1 (1) pT2mN0M0 147 AFD − 
27 F/46 PD 1/4 G1 (1) pT2N0M0 126 AFD − 
28 F/49 LP 1/2.5 G1 (1) pT2N0M0 57 AFD − 
29 M/59 PD 1/5 G1 (1) pT3N1M1 122 AWD − 
30 M/48 MP 1/1.3 G1 (2) pT1N0M0 28 AFD − 
31 M/42 LP 1/5 G1 (1) pT3N1M1 35 AFD − 
32 F/42 MP 1/2 G2 (5) pT2N0M0 56 AFD − 
33 F/59 PD 1/1.5 G2 (10) pT3N1M0 58 DOD − 
34 F/33 LP 1/3.5 G2 (7) pT2N0M0 47 AFD − 
35 F/44 LP 1/5.5 G1 (2) pT2N1M1 55 AWD − 
36 M/56 LP 1/6 G2 (3) pT3N1M1 46 AWD − 
37 F/50 LP 1/4.5 G2 (5) pT3N1M0 42 AWD − 
38 F/35 EN 1/0.9 G2 (5) pT1N0M0 AFD − 
39 M/65 LP 1/4 G2 (5) pT2N1M0 41 AWD − 
40 F/24 LP >15 3/1.5–3.5 G2 (3) pT2mN0M0 AFD − 
41 M/64 EN 1/2 G1 (2) pT2N0M0 62 AFD − 
42 F/72 MP 1/3 G2 (8) pT2N1M0 67 AWD − 
43 M/67 PD 1/5 G2 (8) pT3N0M0 70 AWD − 
44 F/42 EN 1/1.8 G1 (1) pT1N0M0 63 AFD − 
46 F/57 PD 1/2.3 G1 (2) pT2N0M0 34 AFD − 
47 F/43 MP 1/2.3 G1 (1) pT2N0M0 AFD − 
48 F/71 EN 1/2 G2 (10) pT2N0M0 AFD − 
PatientSex/ageSurgical procedureNumber of mAdTumors number/size, cmWHO grade (% Ki-67)TNMFollow-up, moStatusVHL diseaseFrozen tissue/PCR
F/37 PD 1/5 G1 (1) pT3N1M0 155 AFD 
F/28 PD >15 3/0.7–1.5 G1 (1) pT1mN0M0 170 AFD 
F/34 PD 2/1.2–3.2 G1 (1) pT2mN1M0 116 DOD 
M/39 LP 1/3 G1 (1) pT2N0M0 112 DOC 
M/46 PD >15 4/1–3 G1 (2) pT2mN1M0 125 AWD 
F/49 LP 1/2 G2 (10) pT2N0M0 DOC 
M/26 PD >15 2/2 G1 (1) pT2mN0M0 72 AFD 
F/26 PD >15 3/0.6–2.5 G2 (6) pT2mN0M0 44 AFD 
M/51 PD 3/0.8–4.5 G2 (5) pT3mN0M0 13 DOC 
10 F/33 PD 1/2 G2 (5) pT2N1M0 115 AFD 
11 F/28 LP 1/3 G2 (8) pT2N0M0 97 AFD 
12 M/43 LP >15 4/1–4 G2 (7) pT2mN0M0 96 AFD 
13 F/56 LP 1/3.4 G2 (7) pT2N1M0 99 AFD 
14 M/48 PD 5/0.7–2.5 G2 (8) pT2mN1M0 47 AFD 
15 M/30 PD 1/2.5 G2 (5) pT2N0M0 48 AFD 
16 F/33 PD 2/1.3–3.4 G2 (12) pT2mN0M0 60 AFD 
17 F/50 PD 1/5 G1 (1) pT3N1M0 66 AWD 
18 F/35 PD 1/3.2 G2 (5) pT2N0M0 38 AFD 
19 M/26 PD 2/0.6–2.5 G1 (2) pT2mN1M0 87 AWD − 
20 F/44 PD 1/4.2 G2 (8) pT3N0M0 79 AWD − 
21 F/48 PD 3/0.6–2.5 G2 (10) pT2mN0M0 89 AWD − 
22 F/25 PD 3/0.6–2 G1 (1) pT2mN0M0 137 AFD − 
23 F/25 LP 2/0.6–0.8 G1 (1) pT1mN0M1 168 DOC − 
24 F/48 LP 1/4 G1 (1) pT2N0M0 65 AFD − 
25 M/34 EN 1/2 G2 (3) pT2N0M0 12 AFD − 
26 F/76 TP >10 3/1–4 G1 (1) pT2mN0M0 147 AFD − 
27 F/46 PD 1/4 G1 (1) pT2N0M0 126 AFD − 
28 F/49 LP 1/2.5 G1 (1) pT2N0M0 57 AFD − 
29 M/59 PD 1/5 G1 (1) pT3N1M1 122 AWD − 
30 M/48 MP 1/1.3 G1 (2) pT1N0M0 28 AFD − 
31 M/42 LP 1/5 G1 (1) pT3N1M1 35 AFD − 
32 F/42 MP 1/2 G2 (5) pT2N0M0 56 AFD − 
33 F/59 PD 1/1.5 G2 (10) pT3N1M0 58 DOD − 
34 F/33 LP 1/3.5 G2 (7) pT2N0M0 47 AFD − 
35 F/44 LP 1/5.5 G1 (2) pT2N1M1 55 AWD − 
36 M/56 LP 1/6 G2 (3) pT3N1M1 46 AWD − 
37 F/50 LP 1/4.5 G2 (5) pT3N1M0 42 AWD − 
38 F/35 EN 1/0.9 G2 (5) pT1N0M0 AFD − 
39 M/65 LP 1/4 G2 (5) pT2N1M0 41 AWD − 
40 F/24 LP >15 3/1.5–3.5 G2 (3) pT2mN0M0 AFD − 
41 M/64 EN 1/2 G1 (2) pT2N0M0 62 AFD − 
42 F/72 MP 1/3 G2 (8) pT2N1M0 67 AWD − 
43 M/67 PD 1/5 G2 (8) pT3N0M0 70 AWD − 
44 F/42 EN 1/1.8 G1 (1) pT1N0M0 63 AFD − 
46 F/57 PD 1/2.3 G1 (2) pT2N0M0 34 AFD − 
47 F/43 MP 1/2.3 G1 (1) pT2N0M0 AFD − 
48 F/71 EN 1/2 G2 (10) pT2N0M0 AFD − 

Abbreviations: AFD, alive free of disease; AWD, alive with disease; DOC, dead of other cause; DOD, dead of the disease; EN, tumor enucleation; LP, left pancreatectomy; mAd, microadenomas; MP, median pancreatectomy; PD, pancreaticoduodenectomy; and TP, total pancreatectomy.

Controls consisted of 16 non-VHL patients operated on in Beaujon Hospital for PanNET, either sporadic (n = 14) or in the context of a multiple endocrine neoplasia syndrome type 1 (MEN1; n = 2). The median (range) age of patients at the time of surgery was 53 years (33–72). Surgery consisted of 3 pancreaticoduodenectomy, 6 distal pancreatectomy, 3 median pancreatectomies, and 4 enucleations (Table 1). The tumors in VHL and non-VHL patients were matched for mRNA analysis, according to tumor grade [11 of 18 (61%) tumors of grade G2 in patients with VHL vs. 11 of 16 (68%) tumors of grade G2 in non-VHL patients], Ki-67 index (mean Ki-67, 4.77% in VHL tumors vs. 4.81% in non-VHL tumors), tumors size (mean size, 3.09 cm in VHL tumors vs. 3.11 cm in non-VHL tumors), presence of metastases [7 of 18 (38%) metastatic tumors in patients with VHL vs. 6 of 16 (37%) metastatic tumors in non-VHL patients).

For all patients the diagnosis was established on routine formalin-fixed, paraffin-embedded material [hematoxylin and eosin (H&E) sections]. The following histopathologic data were recorded (Table 1): number and size of the tumors, percentage of cell nuclei stained for Ki-67 (Dako, clone MIB1), grading according to tumor-node-metastasis (TNM) and World health Organization (WHO) 2010 classifications (9, 10). The number of microadenomas (100–5,000 μm) and of macroscopic tumors (>5 mm) according to WHO 2010 criteria were also noted (10). For small microadenomas, the expression of both CA9 and chromogranin A was required to distinguish them from large nonneoplastic islets, as described previously (2). Survival data were available for all patients. The median follow-up was 62 months (range, 2–170 months).

Real time RT-PCR

Frozen material for RT-PCR analysis was available for 18 of 31 VHL tumors and 16 of 16 non-VHL tumors, stored at −80°C (Table 1). In all cases one frozen section stained with H&E was obtained before mRNA extraction and histologic examination confirmed that selected samples contained only tumor tissue (both tumor cells and stroma) of good quality and no normal pancreatic tissue. No microdissection was carried out before mRNA extraction. We compared the expression of 52 target genes (Table 2) in VHL and non-VHL tumors. Total RNA was reverse transcribed before real-time PCR amplification (11). All of the PCR reactions were carried out using the Light Cycler 480 Real-Time PCR System and the LightCycler 480 SYBR Green I Master mix (Roche Diagnostics). Quantitative values were obtained from the crossing point (Cp), number at which the increase in the signal associated with exponential growth of PCR products begins to be detected with Light Cycler 480 System Software (Roche Diagnostics), according to the manufacturer's manuals. The precise amount of total RNA added to each reaction mix (based on optical density) and its quality (i.e., lack of extensive degradation) are both difficult to assess. We therefore also quantified transcripts of 2 endogenous RNA control genes involved in 2 cellular metabolic pathways, namely RPLP0 (also known as 36B4; NM_001002), which encodes human acidic ribosomal phosphoprotein P0, and TBP (Genbank accession NM_003194), which encodes the TATA box-binding protein. Each sample was normalized on the basis of its RPLP0 (or TBP) content. We selected RPLP0 because the prevalence of its transcripts is high and because this gene is widely used as an endogenous control for northern blot analysis (more known as 36B4; ref. 11). We also selected TBP as an endogenous control because the prevalence of its transcripts is moderate as compared with RPLP0, and because there are no known TBP retropseudogenes (retropseudogenes lead to coamplification of contaminating genomic DNA and thus interfere with RT-PCR, despite the use of primers in separate exons). Results, expressed as the N-fold differences in target gene expression relative to the RPLP0 gene, and termed Ntarget, was determined as Ntarget = 2ΔCpsample, where the ΔCp value of the sample was determined by substracting the average Cp value of the target gene from the average Cp value of the RPLP0 gene. The Ntarget values of the samples were subsequently normalized to the smallest amount of target gene mRNA, detectable and quantifiable by real-time quantitative PCR assays based on SYBR Green fluorescence methodology (target gene Ct value = 35; Ntarget value = 1).

Table 2.

List of the 52 target genes studied by qRT-PCR

HIF-related molecules  
 CA9 Carbonic anhydrase IX 
 HIF1A Hypoxia-inducible factor 1, α subunit 
 HIF2A Hypoxia-inducible factor 2, α subunit 
 HMOX Heme oxygenase (decycling) 1 
 GLUT Glucose transporter 
Angiogenesis  
 ANGPT1 Angiopoietin 1 
 ANGPT2 Angiopoietin 2 
 CDH5 VE-cadherin (vascular endothelium) 
 CD34 CD34 molecule 
 EDN1 Endothelin 1 
 EDNRA Endothelin receptor type A 
 LYVE1 Lymphatic vessel endothelial hyaluronan receptor 1 
 VEGFA Vascular endothelial growth factor A 
 VEGFB Vascular endothelial growth factor B 
 VEGFR1 FLT1-vascular endothelial growth factor 
 VEGFR2 KDR-kinase insert domain receptor 
EMT  
 CDH1 Cadherin 1, E-cadherin 
 CDH2 Cadherin 2, N-cadherin 
 CLDN1 Claudin 1 
 CTNNB1 Catenin (cadherin-associated protein), β 1 
 JUP Junction plakoglobin 
 OCLN Occludin 
 VIL2 Ezrin 
 VIM Vimentin 
Metastasis-related genes  
 CXCR4 Chemokine (c-x-c) receptor 4 
 LAMA4 Laminin, α 4 
 MMP2 Matrix metallopeptidase 2 (gelatinase A) 
 MMP9 Matrix metallopeptidase 9 (gelatinase B) 
 PLAT Plasminogen activator tissue 
Growth factors and receptors  
 ERBB1 EGF receptor 
 ERBB2 V-erb-b2 erythroblastic leukemia viral oncogene homolog 2 
 ERBB3 V-erb-b2 erythroblastic leukemia viral oncogene homolog 3 
 IGFBP5 Insulin-like growth factor–binding protein 5 
 IRS1 Insulin receptor substrate 1 
 PDGFB Platelet-derived growth factor β polypeptide 
 PTN Pleiotrophin 
 TGFB1 Transforming growth factor, β 1 
 TGFB2 Transforming growth factor, β 2 
 TNFAIP3 Tumor necrosis factor, α-induced protein 3 
Signaling pathways  
 AKT1 V-akt murine thymoma viral oncogene homolog 1 
 DKK3 Dickkopf homolog 3 (Xenopus laevis
 FRAP1/mtor FK506-binding protein 12-rapamycin associated protein 1 
 GADD45B Growth arrest and DNA damage inducible, β 
 GSK3B Glycogen synthase kinase 3 β 
 PTEN Phosphatase and tensin homolog 
 RPS6KB1 Ribosomal protein S6 kinase-polypeptide 1 
 SPRY1 Sprouty homolog 1, antagonist of FGF signaling 
Cell cycle, DNA reparation  
 CCND1 Cyclin D1 
 CDKN1A Cyclin-dependent kinase inhibitor 1A 
 CDKN2A Cyclin-dependent kinase inhibitor 2A 
 KI67 Antigen Ki-67 
 MSH2 MutS homolog 2, colon cancer, nonpolyposis type 1 
HIF-related molecules  
 CA9 Carbonic anhydrase IX 
 HIF1A Hypoxia-inducible factor 1, α subunit 
 HIF2A Hypoxia-inducible factor 2, α subunit 
 HMOX Heme oxygenase (decycling) 1 
 GLUT Glucose transporter 
Angiogenesis  
 ANGPT1 Angiopoietin 1 
 ANGPT2 Angiopoietin 2 
 CDH5 VE-cadherin (vascular endothelium) 
 CD34 CD34 molecule 
 EDN1 Endothelin 1 
 EDNRA Endothelin receptor type A 
 LYVE1 Lymphatic vessel endothelial hyaluronan receptor 1 
 VEGFA Vascular endothelial growth factor A 
 VEGFB Vascular endothelial growth factor B 
 VEGFR1 FLT1-vascular endothelial growth factor 
 VEGFR2 KDR-kinase insert domain receptor 
EMT  
 CDH1 Cadherin 1, E-cadherin 
 CDH2 Cadherin 2, N-cadherin 
 CLDN1 Claudin 1 
 CTNNB1 Catenin (cadherin-associated protein), β 1 
 JUP Junction plakoglobin 
 OCLN Occludin 
 VIL2 Ezrin 
 VIM Vimentin 
Metastasis-related genes  
 CXCR4 Chemokine (c-x-c) receptor 4 
 LAMA4 Laminin, α 4 
 MMP2 Matrix metallopeptidase 2 (gelatinase A) 
 MMP9 Matrix metallopeptidase 9 (gelatinase B) 
 PLAT Plasminogen activator tissue 
Growth factors and receptors  
 ERBB1 EGF receptor 
 ERBB2 V-erb-b2 erythroblastic leukemia viral oncogene homolog 2 
 ERBB3 V-erb-b2 erythroblastic leukemia viral oncogene homolog 3 
 IGFBP5 Insulin-like growth factor–binding protein 5 
 IRS1 Insulin receptor substrate 1 
 PDGFB Platelet-derived growth factor β polypeptide 
 PTN Pleiotrophin 
 TGFB1 Transforming growth factor, β 1 
 TGFB2 Transforming growth factor, β 2 
 TNFAIP3 Tumor necrosis factor, α-induced protein 3 
Signaling pathways  
 AKT1 V-akt murine thymoma viral oncogene homolog 1 
 DKK3 Dickkopf homolog 3 (Xenopus laevis
 FRAP1/mtor FK506-binding protein 12-rapamycin associated protein 1 
 GADD45B Growth arrest and DNA damage inducible, β 
 GSK3B Glycogen synthase kinase 3 β 
 PTEN Phosphatase and tensin homolog 
 RPS6KB1 Ribosomal protein S6 kinase-polypeptide 1 
 SPRY1 Sprouty homolog 1, antagonist of FGF signaling 
Cell cycle, DNA reparation  
 CCND1 Cyclin D1 
 CDKN1A Cyclin-dependent kinase inhibitor 1A 
 CDKN2A Cyclin-dependent kinase inhibitor 2A 
 KI67 Antigen Ki-67 
 MSH2 MutS homolog 2, colon cancer, nonpolyposis type 1 

Primers for endogenous RNA controls and the 52 target genes were chosen with the assistance of the Oligo 5.0 computer program (National Biosciences). To avoid amplification of contaminating genomic DNA, one of the 2 primers was placed at the junction between 2 exons, if possible. In general, amplicons were between 70 and 120 nucleotides long. For each primer pair, no-template control (NTC) and no-reverse transcriptase control (RT negative) assays were conducted and produced negligible signals (usually >40 in Cp value), suggesting that primer dimer formation and genomic DNA contamination effects were negligible. RNA extraction, cDNA synthesis, and PCR reactions have been described previously (12).

Immunohistochemical analysis

Tissue samples.

Tissue microarray (TMA) blocks were constructed from representative blocks from the 31 patients with VHL and the 16 non-VHL patients. For each case at least one macrotumor, including the greatest one, was taken for the TMA (1–4 macrotumors per patient). The construction of these TMA was conducted using a tissue arrayer (Beecher Instruments) and each macrotumor specimen was represented by four 1-mm cores in the TMA block. In case of multiple tumors, the largest one was taken into account for the statistical analysis. If neuroendocrine microadenomatosis was associated, lesions >1 mm were also taken in the TMA (1–4 microadenomas per patient). In total, 3 blocks of TMA were constructed (including 50 macrotumors and 22 microadenomas). In addition to the TMA, we analyzed the normal islets in the pancreas located at a distance from the tumor in 10 of the sporadic patients.

Immunohistochemical procedure

The blocks were cut in 3-μm sections and were immunolabeled with antibodies directed against pVHL (FL181; Santa Cruz Biotechnology), CA9 (polyclonal; Novus Biological), CD34 (QBEND10; Immunotech), cyclin D1 (SP4; Thermo Scientific), vimentin (V9; Dako), E-cadherin (polyclonal; Santa Cruz Biotechnology), occludin (polyclonal; Zymed), and VE-cadherin (F-8; Santa Cruz Biotechnology).

Immunohistochemistry was carried out using an automated immunohistochemical stainer according to the manufacturer's guidelines (Streptavidin-peroxidase with an automate Ventana Benchmark, Ventana). Immunostaining of paraffin sections was conducted after dewaxing and rehydrating slides. Antigen retrieval was conducted by pretreatment with high temperature. Substitution of the primary antibody with PBS was used as a negative control.

Staining evaluation

pVHL, CA9, cyclin D1, vimentin, E-cadherin, and occludin were evaluated by calculating a score obtained by multiplying the intensity (negative, 0; weak, 1; moderate, 2; and strong, 3) by the percentage of stained cells. The pattern of expression (cytoplasmic, membranous, and nuclear) was noted. All cores were evaluated separately and a mean score was calculated for each tumor. Tumors with a score greater than the median value were considered at high protein expression. The heterogeneity of the distribution of the scores was analyzed in scatter plots.

CD34 and VE-cadherin staining allowed evaluation of the microvascular density (MVD). The cores were evaluated separately at high magnification (× 25 objective; area of a core: 0.78 mm2). In each tumor, the core with the highest count was documented. Tumors with an MVD greater than the median value were considered at high MVD. The heterogeneity of the distribution of the MVD evaluated by CD34 and VE-cadherin was analyzed in scatter plots.

Statistical analysis

Comparison of immunohistochemical characteristics between the groups were conducted using the Kruskal–Wallis test for continuous data and the χ2 test or the Fisher exact test for categorical data. Levels of gene mRNA expression between patients with or without a VHL mutation were compared using the Mann–Whitney U test and a ROC (receiver operating characteristics) analysis. The association between histoprognostic factors and levels of gene mRNA expression was searched in the patients with VHL by using the Mann–Whitney U and Spearman tests. Data were analyzed with the SAS 9.1 statistical software for Windows (SAS Institute Inc.). All statistical tests were 2-sided. The critical level of statistical significance was set at P < 0.05.

mRNA expression of the 52 genes in VHL versus non-VHL tumors

Genes upregulated in VHL tumors.

Nineteen (36%) of the 52 genes were significantly upregulated in the tumors in the patients with VHL (Table 3). The upregulated genes were directly related to HIF signaling (CA9/carbonic anhydrase IX; HIF2A/hypoxia-inducible factor 2 α subunit; GLUT1/glucose transporter 1), angiogenesis (CDH5/VE-cadherin; VEGFR1/vascular endothelial growth factor; EDNRA/endothelin receptor type A; ANGPT2/angiopoietin 2; CD34; VEGFR2/kinase insert domain receptor; VEGFA/vascular endothelial growth factor A; ANGPT1/angiopoietin 1), the processes of EMT (VIM/vimentin) and metastasis (LAMA4/lamininα4; CXCR4/chemokine c-x-c receptor 4), growth factors and receptors (PDGFB/platelet-derived growth factor β polypeptide; IRS1/insulin receptor substrate 1; ERBB1/epidermal growth factor receptor), and cell cycle (CCND1/cyclin D1; CDKN2A/cyclin-dependent kinase inhibitor 2A). We obtained the same 19 genes when we compared the VHL tumors with the group of non-VHL/non-NEM1 (n = 14) tumors.

Table 3.

List of the significantly upregulated genes in VHL versus non-VHL patients

Gene symbolsGene definitionTumors in VHL patientsa (n = 18)Tumors in non-VHL patientsa (n = 16)PbROC AUC
HIF-related molecules 
 CA9 Carbonic anhydrase IX 480 (1.5–1,907) 14 (0–502) 0.00095 0.843 
 HIF2A Hypoxia-inducible factor 2, α subunit 2,393 (319–7,325) 593 (183–14,081) 0.0012 0.835 
 GLUT Glucose transporter 1 448 (72–2,423) 138 (20–558) 0.005 0.781 
Angiogenesis 
 CDH5 VE-cadherin (vascular endothelium) 308 (144–1,151) 83 (18–768) 0.00027 0.878 
 VEGFR1 FLT1-vascular endothelial growth factor 716 (175–2,267) 235 (64–1,117) 0.0003 0.865 
 EDNRA Endothelin receptor type A 213 (33–521) 38 (0–290) 0.0003 0.861 
 ANGPT2 Angiopoietin 2 148 (41–2,844) 26 (0–450) 0.00048 0.863 
 CD34 CD34 molecule 105 (29–433) 35 (17–114) 0.0009 0.833 
 VEGFR2 KDR-kinase insert domain receptor 275 (111–593) 122 (34–342) 0.005 0.781 
 VEGFA Vascular endothelial growth factor A 2,688 (734–8,922) 1,118 (181–4,847) 0.008 0.767 
 ANGPT1 Angiopoietin 1 0.19 (0–2.96) 0 (0–0.61) 0.03 0.725 
EMT 
 VIM Vimentin 7,047 (1,840–22,231) 2,110 (19–31,838) 0.00055 0.859 
Metastasis-related genes 
 LAMA4 Laminin α 4 447 (128–1,459) 176 (47–930) 0.0019 0.813 
 CXCR4 Chemokine (c-x-c) receptor 4 90 (19–257) 38 (16–1,157) 0.021 0.733 
Growth factors and receptors 
 PDGFB Platelet-derived growth factor beta polypeptide 369 (55–1,050) 67 (21–2,485) 0.00048 0.863 
 IRS1 Insulin receptor substrate 1 209 (65–472) 134 (27–3,092) 0.011 0.765 
 ERBB1 Epidermal growth factor receptor (EGFR) 259 (10–1,388) 124 (3.8–4,063) 0.016 0.759 
Cell cycle 
 CCND1 Cyclin D1 2,515 (569–11,322) 559 (336–12,386) 0.0009 0.843 
 CDKN2A Cyclin-dependent kinase inhibitor 2A 2.4 (0–6) 0.44 (0–3.38) 0.003 0.799 
Gene symbolsGene definitionTumors in VHL patientsa (n = 18)Tumors in non-VHL patientsa (n = 16)PbROC AUC
HIF-related molecules 
 CA9 Carbonic anhydrase IX 480 (1.5–1,907) 14 (0–502) 0.00095 0.843 
 HIF2A Hypoxia-inducible factor 2, α subunit 2,393 (319–7,325) 593 (183–14,081) 0.0012 0.835 
 GLUT Glucose transporter 1 448 (72–2,423) 138 (20–558) 0.005 0.781 
Angiogenesis 
 CDH5 VE-cadherin (vascular endothelium) 308 (144–1,151) 83 (18–768) 0.00027 0.878 
 VEGFR1 FLT1-vascular endothelial growth factor 716 (175–2,267) 235 (64–1,117) 0.0003 0.865 
 EDNRA Endothelin receptor type A 213 (33–521) 38 (0–290) 0.0003 0.861 
 ANGPT2 Angiopoietin 2 148 (41–2,844) 26 (0–450) 0.00048 0.863 
 CD34 CD34 molecule 105 (29–433) 35 (17–114) 0.0009 0.833 
 VEGFR2 KDR-kinase insert domain receptor 275 (111–593) 122 (34–342) 0.005 0.781 
 VEGFA Vascular endothelial growth factor A 2,688 (734–8,922) 1,118 (181–4,847) 0.008 0.767 
 ANGPT1 Angiopoietin 1 0.19 (0–2.96) 0 (0–0.61) 0.03 0.725 
EMT 
 VIM Vimentin 7,047 (1,840–22,231) 2,110 (19–31,838) 0.00055 0.859 
Metastasis-related genes 
 LAMA4 Laminin α 4 447 (128–1,459) 176 (47–930) 0.0019 0.813 
 CXCR4 Chemokine (c-x-c) receptor 4 90 (19–257) 38 (16–1,157) 0.021 0.733 
Growth factors and receptors 
 PDGFB Platelet-derived growth factor beta polypeptide 369 (55–1,050) 67 (21–2,485) 0.00048 0.863 
 IRS1 Insulin receptor substrate 1 209 (65–472) 134 (27–3,092) 0.011 0.765 
 ERBB1 Epidermal growth factor receptor (EGFR) 259 (10–1,388) 124 (3.8–4,063) 0.016 0.759 
Cell cycle 
 CCND1 Cyclin D1 2,515 (569–11,322) 559 (336–12,386) 0.0009 0.843 
 CDKN2A Cyclin-dependent kinase inhibitor 2A 2.4 (0–6) 0.44 (0–3.38) 0.003 0.799 

Abbreviation: AUC, area under curve.

aMedian and range of gene mRNA levels. The mRNA levels indicated show the abundance of the target relative to the endogenous control (RPLP0). Similar results were obtained with a second endogenous control, TBP.

bMann–Whitney U test.

Genes downregulated in VHL tumors.

Three (6%) of the 52 genes were significantly downregulated in the tumors of the patients with VHL (Table 4). These genes were involved in EMT process (OCLN/occludin) and signaling pathway (RPS6KB1/ribosomal protein S6 kinase-polypeptide 1; GADD45B/growth arrest and DNA damage inducible β). Two of these 3 genes were confirmed when we compared the VHL tumors with the group of non-VHL/non-NEM1 (n = 14) tumors (only GADD45B did not reach significance any more with this analysis, P = 0.13).

Table 4.

List of the significantly downregulated genes in VHL versus non-VHL patients

Gene symbolsGene definitionVHL+ patientsa (n = 18)VHL patientsa (n = 16)PbROC AUC
EMT 
 OCLN Occludin 41 (17–501) 129 (23–1,280) 0.003 0.198 
Signaling pathway 
 RPS6KB1 Ribosomal protein S6 kinase-polypeptide 1 13 (3–34) 21 (11–120) 0.005 0.219 
 GADD45B Growth arrest and DNA damage inducible, β 214 (34–5,094) 466 (12–5,785) 0.01 0.333 
Gene symbolsGene definitionVHL+ patientsa (n = 18)VHL patientsa (n = 16)PbROC AUC
EMT 
 OCLN Occludin 41 (17–501) 129 (23–1,280) 0.003 0.198 
Signaling pathway 
 RPS6KB1 Ribosomal protein S6 kinase-polypeptide 1 13 (3–34) 21 (11–120) 0.005 0.219 
 GADD45B Growth arrest and DNA damage inducible, β 214 (34–5,094) 466 (12–5,785) 0.01 0.333 

Abbreviation: AUC, area under curve.

aMedian and range of gene mRNA levels. The mRNA levels indicated show the abundance of the target relative to the endogenous control (RPLP0). Similar results were obtained with a second endogenous control, TBP.

bMann–Whitney U test.

Correlations between upregulated genes and histopathologic data in patients with VHL.

Among the 19 upregulated genes, we found that high LAMA4, EDNRA, and PDGFA expression correlated with high pT (P = 0.04, P = 0.02, and P = 0.05, respectively) and that high HIF1A expression correlated with greater tumor size (P = 0.01).

Immunohistochemical expression of selected genes in tumors, microadenomas, and normal islets

The scores (range, mean) in VHL tumors, non-VHL tumors, and microadenomas are given in Table 5. The homogeneity of the distribution of the results is presented in scatter plots for each molecule analyzed (Fig. 1).

Figure 1.

Scatter plots showing the distribution of the immunohistochemical results. The distribution of the scores is presented for all molecules tested [pVHL, CA9, CD34, cyclin D1 (c, cytoplasmic; n, nuclear staining), vimentin, E-cadherin, occludin, VE-cadherin] in VHL (Δ) and non-VHL (•) tumors.

Figure 1.

Scatter plots showing the distribution of the immunohistochemical results. The distribution of the scores is presented for all molecules tested [pVHL, CA9, CD34, cyclin D1 (c, cytoplasmic; n, nuclear staining), vimentin, E-cadherin, occludin, VE-cadherin] in VHL (Δ) and non-VHL (•) tumors.

Close modal
Table 5.

Results of immunohistochemical staining in VHL tumors, non-VHL tumors, and microadenomas

VHL tumorsNon-VHL tumorsMicroadenomas
Score range/meanScore range/meanScore range/mean
pVHL (c) 25–200/112.4 100–300/238.5 0–125/95 
CA9 (m) 0–300/188 0–125/20.4 0–300/147 
CD34 (MVD) 18–210/94.8 10–164/47.2 24–244/122 
Cyclin D1 (c) 0–200/47 0–100/7 0–100/31 
Cyclin D1 (n) 0–250/133.6 0–200/52.4 0–270/135 
Vimentin (c) 0–100/82.8 0–67/16.4 0–100/86 
E-cadherin (m) 66–300/198.5 200–300/266.6 100–200/175 
Occludin (c) 0–200/65.5 100–300/238.6 0–100/64 
VE-cadherin (MVD) 3–189/68.6 1–164/40.7 5–219/95 
VHL tumorsNon-VHL tumorsMicroadenomas
Score range/meanScore range/meanScore range/mean
pVHL (c) 25–200/112.4 100–300/238.5 0–125/95 
CA9 (m) 0–300/188 0–125/20.4 0–300/147 
CD34 (MVD) 18–210/94.8 10–164/47.2 24–244/122 
Cyclin D1 (c) 0–200/47 0–100/7 0–100/31 
Cyclin D1 (n) 0–250/133.6 0–200/52.4 0–270/135 
Vimentin (c) 0–100/82.8 0–67/16.4 0–100/86 
E-cadherin (m) 66–300/198.5 200–300/266.6 100–200/175 
Occludin (c) 0–200/65.5 100–300/238.6 0–100/64 
VE-cadherin (MVD) 3–189/68.6 1–164/40.7 5–219/95 

Abbreviations: MVD: microvessel density (vessels/mm2); c, m, and n: cytoplasmic, membranous, and nuclear immunohistochemical staining, respectively.

  • pVHL: Low cytoplasmic expression was correlated with VHL disease (P < 0.0001); the 19 upregulated and 3 downregulated genes in VHL tumors correlated with lower and higher expression of pVHL, respectively (P < 0.05 for all the 22 genes). There was no statistical difference between the expression in tumors and microadenomas in the patients with VHL. In the normal non-VHL islets, the cytoplasmic score ranged from 200 to 300.

  • CA9: High membranous expression correlated with VHL disease (P ≤ 0.0001; Fig. 2A and B). There was no statistical difference between the expression in tumors and microadenomas in patients with VHL. Islets in non-VHL pancreas did not express CA9.

  • CD34: High MVD correlated with VHL disease (P = 0.0008; Fig. 2C and D). The MVD was comparable when assessed by CD34 or VE-cadherin staining (P > 0.05).

  • Cyclin D1: The staining was cytoplasmic or nuclear. High cytoplasmic and nuclear cyclin D1 expressions were both correlated with VHL disease (P = 0.01 and P = 0.001, respectively; Fig. 2E and F). There was no statistical difference between the expression in tumors and microadenomas in patients with VHL. Islets in non-VHL pancreas did not express Cyclin D1.

  • Vimentin: High cytoplasmic expression was correlated with VHL disease (P ≤ 0.0001; Fig. 3A and B). There was no statistical difference between the expression in tumors and microadenomas in the patients with VHL. Islets in non-VHL pancreas did not expressed Vimentin.

  • E-cadherin: Lower membranous expression was correlated with VHL disease (P = 0.0007; Fig. 3C and D). There was no statistical difference between the expression in tumors and microadenomas in the patients with VHL. In the normal non-VHL islets, the score ranged from 150 to 250.

  • Occludin: The staining was mainly cytoplasmic with variable membranous enhancement. High expression was correlated with absence of VHL disease (P ≤ 0.0001; Fig. 3E and F). There was no statistical difference between the expression in tumors and microadenomas in the patients with VHL. In the normal non-VHL islets the cytoplasmic, focally membranous staining score ranged from 100 to 150.

  • VE-cadherin: High MVD correlated with VHL disease (P = 0.04). The MVD was comparable when assessed by CD34 or VE-cadherin staining (P > 0.05).

Figure 2.

Comparison of immunohistochemistry (CA9, CD34, and cyclin D1) on TMA sections in PanNET of VHL (A, C, and E) and non-VHL (B, D, and F) patients. Expression of CA9 is membranous, very strong, and diffuse in the VHL tumors (A) and negative in all tumor cells in non-VHL patients (B). This section also shows a few normal ducts strongly highlighted by CA9, as positive internal control (B). The CD34 staining identifies numerous capillaries and a high MVD in the VHL tumors (C) as compared with the sporadic ones (D). High cytoplasmic and nuclear cyclin D1 expression is detected in VHL tumors (E) in comparison with a light expression in the sporadic ones (F). Original magnifications: × 100 (scale bar = 50 μm).

Figure 2.

Comparison of immunohistochemistry (CA9, CD34, and cyclin D1) on TMA sections in PanNET of VHL (A, C, and E) and non-VHL (B, D, and F) patients. Expression of CA9 is membranous, very strong, and diffuse in the VHL tumors (A) and negative in all tumor cells in non-VHL patients (B). This section also shows a few normal ducts strongly highlighted by CA9, as positive internal control (B). The CD34 staining identifies numerous capillaries and a high MVD in the VHL tumors (C) as compared with the sporadic ones (D). High cytoplasmic and nuclear cyclin D1 expression is detected in VHL tumors (E) in comparison with a light expression in the sporadic ones (F). Original magnifications: × 100 (scale bar = 50 μm).

Close modal
Figure 3.

Immunohistochemical expression on TMA sections of 3 markers of EMT, vimentin, E-cadherin, and occludin in PanNET of VHL (A, C, and E) and non-VHL (B, D, and F) patients. In patients with VHL vimentin shows a strongly cytoplasmic expression in tumor cells (A) as compared with non-VHL tumors where the staining is only detected in the tumor stroma around vessels and negative in all tumor cells (B). Membranous staining of E-cadherin is faint in VHL tumors (C) whereas there is a preservation of the normal membranous staining at cell–cell borders in the sporadic ones (D). Occludin is not detected in tumor cells and only present in endothelial cells in VHL tumors (E) in contrast to sporadic ones (F). Original magnifications: × 100 (scale bar = 50 μm).

Figure 3.

Immunohistochemical expression on TMA sections of 3 markers of EMT, vimentin, E-cadherin, and occludin in PanNET of VHL (A, C, and E) and non-VHL (B, D, and F) patients. In patients with VHL vimentin shows a strongly cytoplasmic expression in tumor cells (A) as compared with non-VHL tumors where the staining is only detected in the tumor stroma around vessels and negative in all tumor cells (B). Membranous staining of E-cadherin is faint in VHL tumors (C) whereas there is a preservation of the normal membranous staining at cell–cell borders in the sporadic ones (D). Occludin is not detected in tumor cells and only present in endothelial cells in VHL tumors (E) in contrast to sporadic ones (F). Original magnifications: × 100 (scale bar = 50 μm).

Close modal

In this article, we have identified a set of genes differentially expressed in PanNET comparing VHL and sporadic patients, mainly involved in angiogenesis, but also in several pathways related to cell cycle and signaling growth factors, extracellular matrix remodeling, and EMT. The up- or downregulation of these genes was significantly correlated with the protein (pVHL) expression status in these tumors. In VHL disease, mutation in the gene leads to considerable decrease of pVHL expression, but we also found that a small subset of sporadic PanNET expressed low levels of pVHL, which could represent somatic VHL alterations, as previously reported (13).

It is well known that in VHL disease, mutation in the VHL gene results in increased expression of HIF, which regulates many angiogenic pathways (14). This is in accordance with the fact that all the tumors developed in patients with VHL, including PanNET, are highly vascularized (2, 15). CD34 and VE-cadherin (cadherin-5) expression is related to tumor angiogenesis (16). We found strong mRNA and protein expression of these molecules in VHL tumors and microadenomas, as we analyzed both tumor cells and their stroma in our samples. VEGF is an important target of HIF and a well-known potent mediator of angiogenesis, overexpressed in VHL associated tumors (17–19). Our results showing a strong mRNA expression of VEGF-A and of its receptors VEGFR1 and VEGFR2 in VHL PanNET are in accordance with those reported in other tumor types associated to VHL disease, suggesting VEGF as a prime candidate for a therapeutic target (20–22). Interestingly, the use of sunitinib, a potent tyrosine kinase inhibitor of VEGF pathway, may have value in the treatment of VHL-related tumors including pheochromocytoma (23). In the presence of VEGF, both angiopoietin 1 and 2 are capable of augmenting angiogenesis (24–27). This parallels well with our results, showing high amounts of both angiopoietin 1 and 2 mRNA in VHL PanNET, as previously observed in renal clear cell carcinomas with alteration of the VHL protein (28). Our results support targeting this system as an effective antiangiogenic therapy in such tumors. Endothelin 1 is a hypoxia-inducible HIF-1 target gene capable of stimulating proliferation and migration, inducing VEGF production and increasing the expression of proto-oncogenes (29–31). We found higher expression of endothelin receptor type A in VHL tumors, which suggests that the regulation of the endothelin pathway is probably highly specific in PanNET in the clinical setting of VHL disease.

EMT permits epithelial cells to acquire mesenchymal like properties via disruption of intercellular adhesion. This process is linked to tumor invasiveness, aggressiveness, and drug resistance (5). Among proteins involved in EMT we found deregulation of vimentin, which is the main intermediate filament of mesenchymal cells. It is upregulated in EMT and is associated with the acquisition of invasion (32). We confirmed strong upregulation of vimentin in PanNET in patients with VHL at both the protein and RNA levels. Vimentin has been shown to be upregulated in VHL-deficient cells as well as in renal clear cell carcinomas and hemangioblastomas in VHL disease (7, 33–35). Transmembrane proteins also play a role in EMT, such as E-cadherin, crucial for the maintenance of structural integrity of epithelia. Its downregulation is a key molecular change in EMT, a process known as “cadherin switch” (5, 36). Lower expression of E-cadherin in VHL tumors at the protein but not at the mRNA level is in favor of a posttranscriptional mechanism of regulation (5). In addition to downregulation of adherens junctions in EMT, tight junctions are involved in this process in in vivo VHL defective sporadic renal clear cell carcinoma, by both HIF-dependent and -independent mechanisms (6). VHL loss of function has a negative effect on the mRNA and protein expression of the tight junction component occludin in PanNET, and also in microadenomas, such as for E-cadherin. Junctions disruption could therefore contribute to tumorigenesis, from the earliest premalignant lesions. This is in accordance with the fact that downregulation of these cell adhesion molecules is involved in malignant transformation in other tumor types, but we found no correlation with clinicopathologic data in our series (37).

Metastasis-related genes are involved in tumor progression. By expressing CXCR4 chemokine receptor, human tumors acquire properties to invade tissue barriers, migrate to secondary organs, and form metastases (38) Loss of pVHL function causes increased expression of CXCR4 (39–41). In accordance with these data, we found strong mRNA expression of CXCR4 in VHL tumors and similar results have been reported in other types of tumors developed in patients with VHL (40, 41). It has recently been shown that CXCR4 triggers expression of metastasis-associated matrix metalloproteinase (MMP) 2/MMP 9, but no difference was found in MMP 2 or MMP 9 mRNA expression between VHL and sporadic PanNET (39). Overexpression of laminin α4 chain has been suggested to correlate with malignancy, migration, and metastatic capacity in different tumor types (42–44). The higher expression of this molecule in our VHL tumors is in line with the earlier discussion on EMT and with the higher risk of malignant course of PanNET in patients with VHL disease (4, 45).

Target genes involved in cell trafficking or acting as growth factors or receptors previously implicated in sporadic NET carcinogenesis and that may represent useful chemotherapeutic targets were also explored. Recent articles have reported high levels of PDGFRB and EGFR in aggressive PanNET (8, 46, 47). Our study reveals that both PDGFRB and the EGFR (ERBB1) are expressed at higher levels in VHL tumors, which supports further investigation of specific targeted anti-PDGFR and EGFR treatment in patients with VHL presenting with a PanNET. Insulin receptor substrate 1 (IRS1), an important mediator in insulin-like growth factor1/insulin signaling, is an adaptor molecule in signal transduction that play a role in development of PanNET (48–51). Interestingly, IRS1 was expressed at higher levels in VHL tumors. GADD45β, which regulates apoptosis, has previously been implicated in the pathogenesis of malignant PanNET (8). Our results point out that it is overexpressed in sporadic tumors and, in contrast, probably not involved in VHL tumorigenesis.

In conclusion, PanNET that develop in the clinical setting of VHL disease express a specific set of genes detected from the very early stages of microadenomas, which lead to consider them as a distinctive subtype of tumors. These specific genes are involved in several metabolic pathways, mainly related to angiogenesis, EMT, and extracellular matrix remodeling, but also to cell signaling. The number of targets regulated directly or indirectly by VHL is likely to be large in PanNET and many but not all are regulated by the HIF system which makes a dominant contribution to alterations in transcription in VHL PanNET.

No potential conflicts of interest were disclosed.

The authors thank the following pathologists, whose names do not appear among the authors, who provided cases and pathological data for this study (*pathologists members of the TENpath network): G. Belleannée* (Bordeaux), S. Casnedi* (Strasbourg), A. Chevallier (Nice), A. Gomez* (Toulouse), P. Guinebretière (Saint-Denis), M.-F. Heymann* (Nantes), C. Laboisse* (Nantes), G. Monges* (Marseille). They are grateful to Sylvie Mosnier, Sandrine Couroble and Véronique Albano for their excellent technical assistance.

Anne Couvelard received a grant from the «GTE: Groupe d'Etude des Tumeurs Endocrines».

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Richard
S
,
Graff
J
,
Lindau
J
,
Resche
F
. 
von Hippel-Lindau disease
.
Lancet
2003
;
36
:
2059
67
.
2.
Périgny
M
,
Hammel
P
,
Corcos
O
,
Larochelle
O
,
Giraud
S
,
Richard
S
, et al
Pancreatic endocrine microadenomatosis in patients with von hippel-Lindau disease: characterization by VHL/HIF pathway proteins expression
.
Am J Surg Pathol
2009
;
33
:
739
48
.
3.
Lubensky
IA
,
Pack
S
,
Ault
D
,
Vortmeyer
AO
,
Libutti
SK
,
Choyke
PL
, et al
Multiple neuroendocrine tumors of the pancreas in von Hippel-lindau disease patients: histopathological and molecular genetic analysis
.
Am J Pathol
1998
;
153
:
223
31
.
4.
Corcos
O
,
Couvelard
A
,
Giraud
S
,
Vullierme
MP
,
O'Toole
D
,
Rebours
V
, et al
Endocrine pancreatic tumors in von Hippel-Lindau disease: clinical, histological, and genetic features
.
Pancreas
2008
;
37
:
85
93
.
5.
Guarino
M
,
Rubino
B
,
Ballabio
G
. 
The role of epithelial-mesenchymal transition in cancer pathology
.
Pathology
2007
;
39
:
305
18
.
6.
Harten
SK
,
Shukla
D
,
Barod
R
,
Hergovich
A
,
Balda
MS
,
Matter
K
, et al
Regulation of renal epithelial tight junctions by the von Hippel-Lindau tumor suppressor gene involves occludin and claudin 1 and is independent of E-cadherin
.
Mol Biol Cell
2009
;
20
:
1089
101
.
7.
Wykoff
CC
,
Pugh
CW
,
Maxwell
PH
,
Harris
AL
,
Ratcliffe
PJ
. 
Identification of novel hypoxia dependent and independent target genes of the von Hippel-Lindau (VHL) tumor suppressor by mRNA differential expression profiling
.
Oncogene
2000
;
19
:
6297
305
.
8.
Duerr
EM
,
Mizukami
Y
,
Ng
A
,
Xavier
RJ
,
Kikuchi
H
,
Deshpande
V
, et al
Defining molecular classifications and targets in gastroenteropancreatic neuroendocrine tumors through DNA microarray analysis
.
Endocr Relat Cancer
2008
;
15
:
243
56
.
9.
Rindi
G
,
Klöppel
G
,
Alhman
H
,
Caplin
M
,
Couvelard
A
,
de Herder
WW
, et al
TNM staging of foregut (neuro) endocrine tumors: a consensus proposal including a grading system
.
Virchows Arch
2006
;
449
:
395
401
.
10.
Rindi
G
,
Klimstra
DS
,
Arnold
R
,
Klöppel
G
,
Bosman
FT
,
Komminoth
P
et al 
Nomenclature and classification of neuroendocrine neoplasms of the digestive system
.
In:
Bosman
FT
,
Carneiro
F
,
Hruban
RH
,
Theise
ND
,
editors
. 
World Health Organization Classification Tumors. Pathology and genetics of tumors of the digestive system
.
Lyon, France
:
IARC Press
; 
2010
.
p.
13
4
.
11.
Bièche
I
,
Lerebours
F
,
Tozlu
S
,
Espie
M
,
Marty
M
,
Lidereau
R
. 
Molecular profiling of inflammatory breast cancer: identification of a poor-prognosis gene expression signature
.
Clin Cancer Res
2004
;
10
:
6789
95
.
12.
Bieche
I
,
Parfait
B
,
Le Doussal
V
,
Olivi
M
,
Rio
MC
,
Lidereau
R
, et al
Identification of CGA as a novel oestrogen receptor-responsive gene in breast cancer: an outstanding candidate marker to predict the response to endocrine therapy
.
Cancer Res
2001
;
61
:
1652
8
.
13.
Schmitt
AM
,
Schmid
S
,
Rudolph
T
,
Anlauf
M
,
Prinz
C
,
Klöppel
G
, et al
VHL inactivation is an important pathway for the development of malignant sporadic pancreatic endocrine tumors
.
Endocr Relat Cancer
2009
;
16
:
1219
27
.
14.
Kaelin
WG
 Jr
. 
The von Hippel-Lindau tumor suppressor protein: O2 sensing and cancer
.
Nat Rev Cancer
2008
;
8
:
865
73
.
15.
Couvelard
A
,
O'Toole
D
,
Turley
H
,
Leek
R
,
Sauvanet
A
,
Degott
C
, et al
Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumors: negative correlation of microvacular density and VEGF expression with tumor progression
.
Br J Cancer
2005
;
92
:
94
101
.
16.
Berx
G
,
van Roy
F
. 
Involvement of members of the cadherin superfamily in cancer
.
Cold Spring Harb Perspect Biol
2009
;
1
:
a003129
.
17.
Baldewijns
MM
,
van Vlodrop
IJ
,
Vermeulen
PB
,
Soetekouw
PM
,
van Engeland
M
,
de Bruïne
AP
. 
VHL and HIF signalling in renal cell carcinogenesis
.
J Pathol
2010
;
221
:
125
38
.
18.
Ferrara
N
,
Gerber
HP
,
LeCouter
J
. 
The biology of VEGF and its receptors
.
Nat Med
2003
;
9
:
669
76
.
19.
Wizigmann-Voos
S
,
Breier
G
,
Risau
W
,
Plate
KH
. 
Up-regulation of vascular endothelial growth factor and its receptors in von Hippel-Lindau disease-associated and sporadic hemangioblastomas
.
Cancer Res
1995
;
55
:
1358
64
.
20.
Chan
CC
,
Collins
AB
,
Chew
EY
. 
Molecular pathology of eyes with von Hippel-Lindau (VHL) Disease: a review
.
Retina
2007
;
27
:
1
7
.
21.
Vortmeyer
AO
,
Tran
MG
,
Zeng
W
,
Gläsker
S
,
Riley
C
,
Tsokos
M
, et al
Evolution of VHL tumorigenesis in nerve root tissue
.
J Pathol
2006
;
210
:
374
82
.
22.
Leung
SY
,
Chan
AS
,
Wong
MP
,
Yuen
ST
,
Fan
YW
,
Chung
LP
. 
Expression of vascular endothelial growth factor in von Hippel-Lindau syndrome-associated papillary cystadenoma of the epididymis
.
Hum Pathol
1998
;
29
:
1322
4
.
23.
Jimenez
C
,
Cabanillas
ME
,
Santarpia
L
,
Jonasch
E
,
Kyle
KL
,
Lano
EA
, et al
Use of the tyrosine kinase inhibitor sunitinib in a patient with von Hippel-Lindau disease: targeting angiogenic factors in pheochromocytoma and other von Hippel-Lindau disease-related tumors
.
J Clin Endocrinol Metab
2009
;
94
:
386
91
.
24.
Davis
S
,
Aldrich
TH
,
Jones
PF
,
Acheson
A
,
Compton
DL
,
Jain
V
, et al
Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expression cloning
.
Cell
1996
;
87
:
1161
9
.
25.
Holash
J
,
Maisonpierre
PC
,
Compton
D
,
Boland
P
,
Alexander
CR
,
Zagzag
D
, et al
Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF
.
Science
1999
;
284
:
1994
8
.
26.
Maisonpierre
PC
,
Suri
C
,
Jones
PF
,
Bartunkova
S
,
Wiegand
SJ
,
Radziejewski
C
, et al
Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis
.
Science
1997
;
277
:
55
60
.
27.
Holash
J
,
Wiegand
SJ
,
Yancopoulos
GD
. 
New model of tumor angiogenesis: dynamic balance between vessel regression and growth mediated by angiopoietins and VEGF
.
Oncogene
1999
;
18
:
5356
62
.
28.
Currie
MJ
,
Gunningham
SP
,
Turner
K
,
Han
C
,
Scott
PA
,
Robinson
BA
, et al
Expression of the angiopoietins and their receptor Tie2 in human renal clear cell carcinomas; regulation by the von Hippel-Lindau gene and hypoxia
.
J Pathol
2002
;
198
:
502
10
.
29.
Hu
J
,
Discher
DJ
,
Bishopric
NH
,
Webster
KA
. 
Hypoxia regulates expression of the endothelin-1 gene through a proximal hypoxia-inducible factor-1 binding site on the antisense strand
.
Biochem Biophys Res Commun
1998
;
245
:
894
9
.
30.
Kozawa
O
,
Kawamura
H
,
Hatakeyama
D
,
Matsuno
H
,
Uematsu
T
. 
Endothelin-1 induces vascular endothelial growth factor synthesis in osteoblasts: involvement of p38 mitogen-activated protein kinase
.
Cell Signal
2000
;
12
:
375
80
.
31.
Herman
WH
,
Simonson
MS
. 
Nuclear signaling by endothelin-1. A Ras pathway for activation of the c-fos serum response element
.
J Biol Chem
1995
;
270
:
11654
61
.
32.
Gilles
C
,
Polette
M
,
Mestdagt
M
,
Nawrocki-Raby
B
,
Ruggeri
P
,
Birembaut
P
, et al
Transactivation of vimentin by β-catenin in human breast cancer cells
.
Cancer Res
2003
;
63
:
2658
64
.
33.
Moch
H
,
Schraml
P
,
Bubendorf
L
,
Mirlacher
M
,
Kononen
J
,
Gasser
T
, et al
High-throughput tissue microarray analysis to evaluate genes uncovered by cDNA microarray screening in renal cell carcinoma
.
Am J Pathol
1999
;
154
:
981
6
.
34.
Ding
XH
,
Zhou
LF
,
Tan
YZ
,
Zhao
Y
,
Zhu
JJ
. 
Histologic and histogenetic investigations of intracranial hemangioblastomas
.
Surg Neurol
2007
;
67
:
239
45
.
35.
Liu
L
,
Qian
J
,
Singh
H
,
Meiers
I
,
Zhou
X
,
Bostwick
DG
. 
Immunohistochemical analysis of chromophobe renal cell carcinoma, renal oncocytoma, and clear cell carcinoma. An optimal and practical panel for differential diagnosis
.
Arch Pathol Lab Med
2007
;
131
:
1290
7
.
36.
Boyer
B
,
Vallés
AM
,
Edme
N
. 
Induction and regulation of epithelial-mesenchymal transitions
.
Biochem Pharmacol
2000
;
60
:
1091
9
.
37.
Osanai
M
,
Murata
M
,
Nishikiori
N
,
Chiba
H
,
Kojima
T
,
Sawada
N
. 
Epigenetic silencing of occludin promotes tumorigenic and metastatic properties of cancer cells via modulations of unique sets of apoptosis-associated genes
.
Cancer Res
2006
;
66
:
9125
33
.
38.
Kucia
M
,
Reca
R
,
Miekus
K
,
Wanzeck
J
,
Wojakowski
W
,
Janowska-Wieczorek
A
, et al
Trafficking of normal stem cells and metastasis of cancer stem cells involve similar mechanisms: pivotal role of the SDF-1-CXCR4 axis
.
Stem Cells
2005
;
23
:
879
94
.
39.
Struckmann
K
,
Mertz
K
,
Steu
S
,
Storz
M
,
Staller
P
,
Krek
W
, et al
pVHL co-ordinately regulates CXCR4/CXCL12 and MMP2/MMP9 expression in human clear-cell renal cell carcinoma
.
J Pathol
2008
;
214
:
464
71
.
40.
Staller
P
,
Sulitkova
J
,
Lisztwan
J
,
Moch
H
,
Oakeley
EJ
,
Krek
W
. 
Chemokine receptor CXCR4 downregulated by von Hippel-Lindau tumor suppressor pVHL
.
Nature
2003
;
425
:
307
11
.
41.
Zagzag
D
,
Krishnamachary
B
,
Yee
H
,
Okuyama
H
,
Chiriboga
L
,
Ali
MA
, et al
Stromal cell-derived factor-1α and CXCR4 expression in hemangioblastoma and clear cell-renal cell carcinoma: von Hippel–Lindau loss-of-function induces expression of a ligand and its receptor
.
Cancer Res
2005
;
65
:
6178
88
.
42.
Nagato
S
,
Nakagawa
K
,
Harada
H
,
Kohno
S
,
Fujiwara
H
,
Sekiguchi
K
, et al
Downregulation of laminin alpha4 chain expression inhibits glioma invasion in vitro and in vivo
.
Int J Cancer
2005
;
117
:
41
50
.
43.
Vainionpää
N
,
Lehto
VP
,
Tryggvason
K
,
Virtanen
I
. 
Alpha 4 chain laminins are widely expressed in renal cell carcinomas and have a de-adhesive function
.
Lab Invest
2007
;
87
:
780
91
.
44.
Ljubimova
JY
,
Fugita
M
,
Khazenzon
NM
,
Das
A
,
Pikul
BB
,
Newman
D
, et al
Association between laminin-8 and glial tumor grade, recurrence, and patient survival
.
Cancer
2004
;
101
:
604
12
.
45.
Libutti
SK
,
Choyke
PL
,
Alexander
HR
,
Glenn
G
,
Bartlett
DL
,
Zbar
B
, et al
Clinical and genetic analysis of patients with pancreatic neuroendocrine tumors associated with von Hippel-Lindau disease
.
Surgery
2000
;
128
:
1022
8
.
46.
Fjällskog
ML
,
Lejonklou
MH
,
Oberg
KE
,
Eriksson
BK
,
Janson
ET
. 
Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors
.
Clin Cancer Res
2003
;
9
:
1469
73
.
47.
Bergmann
F
,
Breinig
M
,
Höpfner
M
,
Rieker
RJ
,
Fischer
L
,
Köhler
C
, et al
Expression pattern and functional relevance of epidermal growth factor receptor and cyclooxygenase-2: novel chemotherapeutic targets in pancreatic endocrine tumors?
.
Am J Gastroenterol
2009
;
104
:
171
81
.
48.
White
MF
. 
Insulin signaling in health and disease
.
Science
2003
;
302
:
1710
1
.
49.
Reiss
K
,
Wang
JY
,
Romano
G
,
Furnari
FB
,
Cavenee
WK
,
Morrione
A
, et al
IGF-I receptor signaling in a prostatic cancer cell line with a PTEN mutation
.
Oncogene
2000
;
19
:
2687
94
.
50.
Gibson
SL
,
Ma
Z
,
Shaw
LM
. 
Divergent roles for IRS-1 and IRS-2 in breast cancer metastasis
.
Cell Cycle
2007
;
6
:
631
7
.
51.
Shen
Q
,
Sun
Q
,
Wei
X
,
Dong
J
,
Zhang
R
,
Wu
P
, et al
Generation and characterization of islet cell tumor in pTet-on/pTRE-SV40Tag double-transgenic mice model
.
J Biosci Bioeng
2007
;
104
:
14
21
.