Purpose:p14ARF, p15INK4b, and p16INK4a are tumor suppressor genes that are located closely at 9p21 and are often coinactivated by genetic or epigenetic alterations. Malignant peripheral nerve sheath tumor (MPNST) is a rare sarcoma with poor prognosis. However, the prognostic implications of inactivation of p14ARF, p15INK4b, and p16INK4a in MPNSTs have not been adequately investigated. Here we carried out a genetic, epigenetic, and expression analysis of p14ARF, p15INK4b, and p16INK4a, and clarified the prognostic significance of their inactivation in MPNSTs.

Experimental Design: p14ARF, p15INK4b, and p16INK4a protein expressions were assessed by immunohistochemistry in 129 formalin-fixed samples of MPNST including 85 primary tumors. Thirty-nine samples, for which frozen material was available, were also investigated by Western blotting and quantitative reverse transcription PCR (RT-PCR) to detect p14ARF, p15INK4b, and p16INK4a protein and mRNA expression, and by multiplex real-time PCR, PCR single strand conformation polymorphism and methylation-specific PCR to detect p14ARF, p15INK4b, and p16INK4a gene alterations.

Results: Immunohistochemically decreased expressions of p14ARF, p15INK4b, and p16INK4a were observed in 48%, 54%, and 49% of primary MPNSTs, respectively, and were significantly correlated with their concordant mRNA levels. As for gene alterations, homozygous deletion of CDKN2A was detected in one third of the cases. Inactivation of p14ARF and p16INK4a was associated with poor prognosis by both univariate and multivariate analyses. Furthermore, cases with inactivation of all p14ARF, p15INK4b, and p16INK4a genes showed the worst prognosis in a combined prognostic assessment.

Conclusion: A comprehensive analysis of p14ARF, p15INK4b, and p16INK4a inactivation status provides useful prognostic information in MPNSTs. Clin Cancer Res; 17(11); 3771–82. ©2011 AACR.

Translational Relevance

p14ARF, p15INK4b, and p16INK4a are tumor suppressor genes that are located closely at 9p21 and are often coinactivated by genetic or epigenetic alterations. Malignant peripheral nerve sheath tumor (MPNST) is a soft tissue sarcoma carrying poor prognosis, but the prognostic effect of p14ARF, p15INK4b, and p16INK4a in MPNSTs has not been sufficiently investigated. We carried out a comprehensive analysis of p14ARF, p15INK4b, and p16INK4a in a large number of MPNST samples. The results showed that the inactivations of p14ARF and p16INK4a were independently associated with poor prognosis. The prognostic analysis suggested that p15INK4b provides a backup function for p16INK4a. Cases with inactivation of all 3 genes showed the worst prognosis in a combined prognostic analysis. We propose that a comprehensive analysis of p14ARF, p15INK4b, and p16INK4a provides useful prognostic information in cases of MPNST.

Malignant peripheral nerve sheath tumor (MPNST) is an uncommon soft tissue sarcoma carrying poor prognosis (1). Approximately half of MPNSTs develop from benign neurofibroma in patients with neurofibromatosis type 1 (NF1), whereas the remaining half occur sporadically (2). NF1 is an autosomal dominant neurocutaneous disorder with NF1 gene dysfunction, and characterized by multiple neurofibromas and greatly increased risk of developing MPNST. However, loss of both NF1 alleles is not sufficient for malignant change of neurofibromas, and thus additional genetic alterations are thought to be necessary for the malignant transformation (3–6). To date, however, the detailed molecular events contributing to development of MPNST have been unclear in both NF1-related and sporadic cases.

Clinically, surgical resection is the primary curative treatment of MPNSTs (7). The roles of chemotherapy and radiation therapy are very limited (8). Because unresectable cases are unlikely to be cured, early detection of recurrent or metastatic lesions is of great importance in the follow-up of postoperative patients with MPNST. For the selection of high-risk patients, precise prognostic prediction is important, even though it has not been established yet.

Frequent loss of the CDKN2ACDKN2B locus on chromosome 9p21 has been reported in various kinds of human cancers and sarcomas (9–13). The locus encodes 3 important cell-cycle inhibitory proteins: p14ARF, which is encoded by an alternative reading frame of CDKN2A, p15INK4b, which is encoded by CDKN2B, and p16INK4a, which is encoded by CDKN2A. The p14ARF protein activates the key checkpoint protein p53, thereby inducing either cell-cycle arrest (both in G1 and G2) or apoptosis. Both p15INK4b and p16INK4a are able to induce cell-cycle arrest in G1 by inhibiting the cyclin-dependent kinases CDK4 and CDK6 to inactivate the retinoblastoma (RB1) family of tumor suppressor proteins. The tumor-suppressive functions for p16INK4a and p14ARF have been firmly established through experiments; however, the role of p15INK4b remains controversial because transgenic mice deficient for p15INK4b showed only a very subtle tumor predisposition in an experimental study (14). More recently, it has been shown that p15INK4b functions as a critical tumor suppressor in the absence of p16INK4a in an experimental setting (15), whereas the tumor-suppressive effect of p15INK4b has not been investigated well in clinical tumor specimens.

Several studies have shown that p16INK4a is more frequently inactivated in MPNSTs than in neurofibromas (16–24). On the other hand, the impact of p14ARF and p15INK4b inactivation in MPNSTs has been less investigated. Furthermore, the prognostic influence of p14ARF, p15INK4b, and p16INK4a inactivation in patients with MPNST has not been examined enough.

Here we carried out a comprehensive analysis of the 9p21-related genes p14ARF, p15INK4b, and p16INK4a in a series of 129 paraffin-embedded and 39 snap-frozen specimens from 99 patients with MPNST. The gene alterations and mRNA and protein expressions were studied by multiplex real-time PCR, PCR–single-strand conformation polymorphism (PCR–SSCP), methylation-specific PCR (MSP), quantitative reverse transcription PCR (RT-PCR), Western blotting (WB), and immunohistochemistry (IHC). Finally, the immunohistochemical results were compared with the clinicopathological variables and overall survival of the patients.

Patients and tumors

One hundred twenty-nine paraffin-embedded MPNST specimens from 99 patients were obtained from the collection of soft tissue tumors registered in the Department of Anatomic Pathology, Pathological Sciences, Kyushu University, Japan between 1964 and 2008. The specimens included 85 primary, 39 recurrent, and 5 metastatic tumors. Specimens of the primary tumor were not available from 14 patients who visited our hospital at the time of recurrence. A total of 18 pairs of specimens from the primary and the corresponding first recurrent or metastatic tumors were available for the purpose of comparison. The diagnosis of MPNST was made according to the latest edition of the World Health Organization classification (25) and the textbook of Enzinger and Weiss (26). The clinicopathological characteristics are summarized in Table 1. The 85 primary cases included 45 males and 40 females whose ages ranged from 1 to 88 years (median 47 years). In all, 33 patients were diagnosed with NF1 based on the NIH criteria (27). Tumors were located in the extremities in 38 cases (thigh 18, upper arm 13, lower leg 5, forearm 2), in the trunk wall in 20 cases (back 8, buttock 6, chest wall 4, abdominal wall 2), in the spinal or paraspinal region in 11 cases, in the head and neck in 9 cases, in the retroperitoneum in 5 cases, and in the visceral organs in 2 cases. All primary tumors were treated surgically, accompanied by adjuvant chemotherapy or irradiation in 3 cases. The histological tumor grade was evaluated in all specimens according to the French Federation of Cancer Centers (FNCLCC) grading system (28). As for staging of the primary tumors, the latest American Joint Committee on Cancer (AJCC) staging system was used (29). Follow-up information was available in 82 out of the 85 primary tumor cases. The median follow-up period after surgery was 27 months (range, 3–291 months).

Table 1.

Association between clinicopathological variables and immunohistochemical results of p14ARF, p15INK4b, and p16INK4a

p14ARFp15INK4bp16INK4a
VariableNo. of patientsPreservedDecreasedPPreservedDecreasedPPreservedDecreasedP
Clinical variables (primary cases, n = 85)           
 Age, y           
  <50 46 24 22 0.93 21 25 0.96 22 24 0.58 
  ≥50 39 20 19  18 21  21 18  
 Sex           
  Male 45 26 19 0.24 22 23 0.56 20 25 0.23 
  Female 40 18 22  17 23  23 17  
 NF1           
  Present 33 16 17 0.63 14 19 0.61 15 18 0.45 
  Absent 52 28 24  25 27  28 24  
 Site           
  Extremity 38 20 18 0.98 17 21 0.29 19 19 0.77 
  Trunk wall 20 10 10  12  11  
  Head and neck/  retroperitoneum/  visceral/spine 27 14 13  10 17  15 12  
 Tumor depth           
  Superficial 21 12 0.57 12 0.23 12 0.41 
  Deep 64 32 32  27 37  34 30  
 Tumor size           
  <5cm 24 15 0.21 16 0.015 15 0.17 
  ≥5cm 61 29 32  23 38  28 33  
 Adjuvant therapy           
  Given 0.11 0.59 1.00 
  Not given 82 44 38  37 45  41 41  
 AJCC staging           
  I 26 14 12 0.21 14 12 0.63 14 12 0.81 
  II 41 21 20  16 25  22 19  
  III 14    
  IV    
Pathological variables (all cases, n = 129)           
 Tumor necrosis           
  No necrosis 63 37 26 0.30 39 24 0.17 34 29 0.28 
  <50% 52 23 29  24 28  31 21  
  ≥50% 14    
 Mitotic counts           
  0–9/10HPF 84 45 39 0.11 42 42 0.42 41 43 0.11 
  10–19/10HPF 17 12    
  ≥20/10HPF 28 17 11  18 10  20  
 Ki-67–labeling index           
  0%–9% 31 20 11 0.043 13 18 0.33 17 14 0.94 
  10%–29% 57 32 25  32 25  30 27  
  ≥30% 41 15 26  24 17  23 18  
 Microvessel density           
  <15/HPF 64 29 35 0.13 33 31 0.66 31 33 0.19 
  ≥15/HPF 65 38 27  36 29  39 26  
 Rhabdomyoblastic differentiation           
  Present 16 11 0.074 0.77 0.37 
  Absent 113 62 51  61 52  63 50  
 Epithelioid feature           
  Present 0.11 0.091 0.80 
  Absent 121 65 56  67 54  66 55  
 FNCLCC grading           
  1 41 25 16 0.37 26 15 0.066 20 21 0.68 
  2 59 28 31  25 34  33 26  
  3 29 14 15  18 11  17 12  
p14ARFp15INK4bp16INK4a
VariableNo. of patientsPreservedDecreasedPPreservedDecreasedPPreservedDecreasedP
Clinical variables (primary cases, n = 85)           
 Age, y           
  <50 46 24 22 0.93 21 25 0.96 22 24 0.58 
  ≥50 39 20 19  18 21  21 18  
 Sex           
  Male 45 26 19 0.24 22 23 0.56 20 25 0.23 
  Female 40 18 22  17 23  23 17  
 NF1           
  Present 33 16 17 0.63 14 19 0.61 15 18 0.45 
  Absent 52 28 24  25 27  28 24  
 Site           
  Extremity 38 20 18 0.98 17 21 0.29 19 19 0.77 
  Trunk wall 20 10 10  12  11  
  Head and neck/  retroperitoneum/  visceral/spine 27 14 13  10 17  15 12  
 Tumor depth           
  Superficial 21 12 0.57 12 0.23 12 0.41 
  Deep 64 32 32  27 37  34 30  
 Tumor size           
  <5cm 24 15 0.21 16 0.015 15 0.17 
  ≥5cm 61 29 32  23 38  28 33  
 Adjuvant therapy           
  Given 0.11 0.59 1.00 
  Not given 82 44 38  37 45  41 41  
 AJCC staging           
  I 26 14 12 0.21 14 12 0.63 14 12 0.81 
  II 41 21 20  16 25  22 19  
  III 14    
  IV    
Pathological variables (all cases, n = 129)           
 Tumor necrosis           
  No necrosis 63 37 26 0.30 39 24 0.17 34 29 0.28 
  <50% 52 23 29  24 28  31 21  
  ≥50% 14    
 Mitotic counts           
  0–9/10HPF 84 45 39 0.11 42 42 0.42 41 43 0.11 
  10–19/10HPF 17 12    
  ≥20/10HPF 28 17 11  18 10  20  
 Ki-67–labeling index           
  0%–9% 31 20 11 0.043 13 18 0.33 17 14 0.94 
  10%–29% 57 32 25  32 25  30 27  
  ≥30% 41 15 26  24 17  23 18  
 Microvessel density           
  <15/HPF 64 29 35 0.13 33 31 0.66 31 33 0.19 
  ≥15/HPF 65 38 27  36 29  39 26  
 Rhabdomyoblastic differentiation           
  Present 16 11 0.074 0.77 0.37 
  Absent 113 62 51  61 52  63 50  
 Epithelioid feature           
  Present 0.11 0.091 0.80 
  Absent 121 65 56  67 54  66 55  
 FNCLCC grading           
  1 41 25 16 0.37 26 15 0.066 20 21 0.68 
  2 59 28 31  25 34  33 26  
  3 29 14 15  18 11  17 12  

This study was conducted in accordance with the principles embodied in the Declaration of Helsinki. The study was also approved by the Ethics Committee of Kyushu University (No. 21–137) and conducted according to the Ethical Guidelines for Epidemiological Research enacted by the Japanese Government.

Immunohistochemistry

Immunohistochemical staining was carried out in the same way as described previously (10). Sections were pretreated in a microwave oven at 100°C for 20 minutes before being incubated with monoclonal antibodies of p14ARF (DCS-240, 1:1,000; Sigma-Aldrich), p15INK4b (15P06, 1:50; Abcam), and p16INK4a (F-12, 1:100; Santa Cruz Biotechnology) at 4°C overnight. Normal tonsil tissue or colon cancer tissue was used as an external positive control. As a negative control, the primary antibody was omitted. Immunohistochemical results were judged by 3 investigators (M.E., N.S., and Y.O.), who were blinded to the clinical status of the patients. A consensus judgment was adopted as the proper immunohistochemical result based on the proportion of nuclear-stained tumor cells. When less than 50% of the tumor cells were stained, the expression was considered decreased by reference to the previously published papers (10, 30, 31). The serial sections were also immunostained with anti–Ki-67 antibody (M 7240, 1:100; Dako) and anti-CD31 antibody (JC70A, 1:20; Dako) using the standard procedure. The Ki-67–labeling index and microvessel density (MVD) were calculated as described previously (32, 33).

Snap-frozen samples and WB analysis

Thirty-nine snap-frozen samples from 28 primary and 11 recurrent or metastatic tumors were obtained from the collection of our department. A total of 5 pairs of samples from the primary and the corresponding first recurrent or metastatic tumors were available for the purpose of comparison. Fresh tumor samples were carefully dissected for the tumors to exclude the surrounding normal tissue, and the samples were immediately frozen in liquid nitrogen and stored at −80°C.

To confirm the concordant immunohistochemical results, the expressions of p14ARF, p15INK4b, and p16INK4a protein were evaluated by WB analysis using snap-frozen samples as previously described (34). A total of 20 μg protein from each sample was used, and incubated with either anti-p14ARF (1:2,000 dilution), -p15INK4b (1:50), or -16INK4a (1:200) antibodies. Anti-human actin mouse monoclonal antibody (1:5,000; Millipore) was used as an internal control. A frozen sample of normal skin tissue was used as an external positive control. Protein levels were standardized by actin, which was assigned an arbitrary level of 10, and the expression signal relative to this was taken as the expression value for each sample.

RNA extraction and quantitative RT-PCR

Total RNA was extracted from 39 frozen samples and a cell line as previously described (35). The MPNST cell line YST-1 (36), which expresses CDKN2A and CDKN2B mRNAs, was used as a positive control. Quantitative RT-PCR was carried out and the results were analyzed using predeveloped TaqMan assay reagents (CDKN2A Hs00233365_m1 and CDKN2B Hs00793225_m1 from Applied Biosystems, Life Technologies) and an ABI Prism 7700 Sequence Detection system (Applied Biosystems) as described previously (37). The standard curve was constructed with serial dilutions of YST-1 cell line samples. The obtained data were standardized by using data of the housekeeping gene, GAPDH (Hs99999905_m1). The final numerical value (V) in each sample was calculated as follows: V = CDKN2A or CDKN2B mRNA value/GAPDH mRNA value.

DNA extraction and analyses

Genomic DNA was isolated from 27 snap-frozen tumor materials by using standard proteinase K digestion and phenol/chloroform extraction as previously described (38).

Multiplex real-time PCR was carried out for detecting homozygous deletions (HD) of the CDKN2A gene. The primer sequences and PCR conditions were the same as those described previously (39). All primers, the TaqMan probes and the TaqMan Universal Master Mix were supplied by Applied Biosystems. A standard curve was constructed with serial dilutions of Human Genomic DNA (Clontech). Distilled water was used as a negative control in each PCR analysis. The CDKN2A/beta-actin ratios less than 40% were judged to represent HD according to the previous reports (39, 40).

Mutational analysis was carried out for exons 1 to 3 of the CDKN2A gene by PCR-SSCP and DNA sequencing. The primer sequences and PCR conditions were the same as those previously described (9). SSCP and the following DNA sequencing were carried out as described previously (41).

Methylation of the CpG island in the promoter region of the p14ARF and p16INK4a genes was determined by MSP. Bisulfite modification and MSP were carried out as described previously, using the same reagents, primers, and PCR conditions (9, 42).

Statistical analysis

The correlations between 2 dichotomous variables were evaluated by using either χ2 test or, when appropriate, Fisher's exact test. The correlations among gene alteration, mRNA level, and protein expression were evaluated by Mann–Whitney U test. The difference in the proportion of p14ARF-, p15INK4b-, or p16INK4a-positive cells between the primary and the recurrent tumors was evaluated by Wilcoxon signed-rank test. Survival curves were calculated with the Kaplan–Meier method, and the differences were compared by the log-rank test. Cox proportional hazards regression analysis was carried out to estimate the hazard ratios for positive risk factors for death. Statistical significance was defined as P < 0.05. Data analysis was carried out with the JMP statistical software package (version 8.0.2; SAS Institute Inc.).

Immunohistochemistry

The representative examples of immunohistochemical staining are shown in Figure 1, and the correlations between p14ARF, p15INK4b, and p16INK4a immunoreactivity and the clinicopathological variables are summarized in Table 1. Almost all neurofibroma cells showed immunoreactivity for p14ARF, p15INK4b, and p16INK4a, respectively, on the 12 paraffin-embedded specimens including a benign neurofibroma area with MPNST. p14ARF expression was decreased in 41of the 85 primary tumors (48%) and 62 of the 129 all specimens (48%). For the total group of 129 cases, decreased expression of p14ARF was frequently observed in the subgroup with a high Ki-67–labeling index (P = 0.043). p15INK4b expression was decreased in 46 of the 85 primary tumors (54%) and 60 of the 129 all specimens (47%). Decreased expression of p15INK4b was associated with large tumor size (more than 5 cm) in both the primary tumors (P = 0.015) and all specimens (P = 0.0003). p16INK4a expression was decreased in 42 of the 85 primary tumors (49%) and 59 of the 129 all specimens (46%). p16INK4a expression showed no significant correlation with any clinicopathological variables.

Figure 1.

Representative examples of immunohistochemical staining. A, nuclear staining of p14ARF in MPNST. B, p14ARF expression in the gradual transition area from neurofibroma to MPNST. Most MPNST cells with plump nuclei were negative for p14ARF (MPNST, bottom right). Most spindle-shaped neurofibroma cells were positive for p14ARF (NF, top left). C, nuclear staining of p15INK4b in MPNST. D, decreased expression of p15INK4b in the MPNST area (MPNST, right). The expression was preserved in vascular endothelial cells and stromal cells in the surrounding normal tissue (N, left). E, nuclear staining of p16INK4a in MPNST. F, decreased expression of p16INK4a in MPNST. The expression was preserved in vascular endothelial cells. G, WB analysis of p16INK4a expression for a comparison between the primary and the corresponding recurrent tumors. The values at the top of the panel indicate the level of p16INK4a expression; this value was calculated relative to the actin intensity, which was assigned a value of 10. The expression level was decreased in the recurrent tumors of cases 1 and 2. A subtle decrease was observed in case 3. Case 4 shows no change, and case 5 shows an increase in p16INK4a expression. Cont.: positive control; P: primary; R: recurrent.

Figure 1.

Representative examples of immunohistochemical staining. A, nuclear staining of p14ARF in MPNST. B, p14ARF expression in the gradual transition area from neurofibroma to MPNST. Most MPNST cells with plump nuclei were negative for p14ARF (MPNST, bottom right). Most spindle-shaped neurofibroma cells were positive for p14ARF (NF, top left). C, nuclear staining of p15INK4b in MPNST. D, decreased expression of p15INK4b in the MPNST area (MPNST, right). The expression was preserved in vascular endothelial cells and stromal cells in the surrounding normal tissue (N, left). E, nuclear staining of p16INK4a in MPNST. F, decreased expression of p16INK4a in MPNST. The expression was preserved in vascular endothelial cells. G, WB analysis of p16INK4a expression for a comparison between the primary and the corresponding recurrent tumors. The values at the top of the panel indicate the level of p16INK4a expression; this value was calculated relative to the actin intensity, which was assigned a value of 10. The expression level was decreased in the recurrent tumors of cases 1 and 2. A subtle decrease was observed in case 3. Case 4 shows no change, and case 5 shows an increase in p16INK4a expression. Cont.: positive control; P: primary; R: recurrent.

Close modal

As for the difference in the IHC results between NF1-related and sporadic MPNSTs, p14ARF expression was decreased in 17 of the 33 NF1-related (52%) and 24 of the 52 sporadic cases (46%). p15INK4b expression was decreased in 19 of the 33 NF1-related (58%) and 27 of the 52 sporadic (52%) cases. p16INK4a expression was decreased in 18 of the 33 NF1-related (55%) and 24 of the 52 sporadic (46%) MPNSTs. There was no significant difference in p14ARF, p15INK4b, and p16INK4a expressions between NF1-related and sporadic MPNSTs.

A comparative analysis of the immunoreactivities for p14ARF, p15INK4b, and p16INK4a was carried out in the 18 pairs of the primary and the corresponding first recurrent or metastatic tumors. A decrease in the proportion of p14ARF-, p15INK4b-, and p16INK4a-immunoreactive cells was observed in 10 (56%), 12 (67%), and 13 (72%) out of the 18 recurrent or metastatic tumors, respectively. There was no significant difference in positivity between the primary and the corresponding recurrent tumors; however, p16INK4a immunoreactivity showed a tendency to be lower in the recurrent or metastatic tumors (P = 0.12).

Western blotting

Proteins of sufficient quality and quantity were successfully extracted from 38 out of the 39 frozen tumor specimens. WB analysis was carried out in 38 specimens using anti-p14ARF, -p15INK4b, and -p16INK4a antibodies. The p14ARF signal was detected in only 4 cases, and 3 of the 4 cases in which the p14ARF signal was detected on WB also presented preserved expression of this protein on IHC (Table 2). The p15INK4b signal was observed in only 3 cases, and all 3 cases in which the p15INK4b signal was detected on WB also presented preserved expression of this protein on IHC (Table 2). WB for the p16INK4a protein showed a sufficient signal intensity to evaluate the relative expression levels in 34 of the 38 samples (Table 2). The relative expression level versus the actin expression level, which was assigned a value of 10, ranged from 0.0 to 26.9 (median 2.1). As for the comparison of the primary and the corresponding recurrent or metastatic tumors, p16INK4a expression levels revealed a decrease in 2 of 5 cases, and a subtle decrease in 1 case (Fig. 1G). The expression levels of p16INK4a by WB corresponded closely to the levels observed by IHC (Table 2).

Table 2.

A comparison of p14ARF, p15INK4b, and p16INK4a expression results by IHC, WB, and qPCR

p14ARFp15INK4bp16INK4a
Case no.IHCWBqPCRIHCWBqPCRIHCWBqPCR
0.00 2.01 17.4 0.00 
0.00 0.9 0.03 2.1 0.00 
0.00 0.01 0.00 
0.00 0.00 2.1 0.00 
0.00 0.00 1.6 0.00 
0.01 0.02 9.6 0.01 
0.02 0.00 0.02 
0.03 0.28 0.4 0.03 
0.03 0.09 0.4 0.03 
10 0.03 0.03 4.8 0.03 
11 0.04 0.03 2.1 0.04 
12 4.7 0.04 0.02 1.9 0.04 
13 0.05 0.13 0.6 0.05 
14 0.05 0.11 0.05 
15 0.05 0.00 3.8 0.05 
16 0.06 0.72 0.06 
17 0.08 0.27 11.4 0.08 
18 0.09 0.00 0.4 0.09 
19 0.10 0.47 0.7 0.10 
20 0.10 0.00 1.5 0.10 
21 0.12 0.46 0.2 0.12 
22 0.14 0.06 9.8 0.14 
23 0.14 0.01 26.9 0.14 
24 0.15 3.5 10.64 12 0.15 
25 0.44 1.50 2.2 0.44 
26 0.52 0.07 2.2 0.52 
27 0.54 1.07 0.1 0.54 
28 0.69 0.09 0.69 
29 1.04 0.16 1.04 
30 1.71 0.05 6.8 1.71 
31 3.08 0.40 3.1 3.08 
32 0.9 4.12 1.65 0.4 4.12 
33 4.29 0.74 4.1 4.29 
34 4.41 1.37 4.41 
35 1.4 4.50 2.2 0.00 8.9 4.50 
36 4.77 4.13 0.4 4.77 
37 9.00 2.28 9.00 
38 n.e. 9.72 n.e. 0.11 n.e. 9.72 
39 3.6 11.23 6.13 2.2 11.23 
p14ARFp15INK4bp16INK4a
Case no.IHCWBqPCRIHCWBqPCRIHCWBqPCR
0.00 2.01 17.4 0.00 
0.00 0.9 0.03 2.1 0.00 
0.00 0.01 0.00 
0.00 0.00 2.1 0.00 
0.00 0.00 1.6 0.00 
0.01 0.02 9.6 0.01 
0.02 0.00 0.02 
0.03 0.28 0.4 0.03 
0.03 0.09 0.4 0.03 
10 0.03 0.03 4.8 0.03 
11 0.04 0.03 2.1 0.04 
12 4.7 0.04 0.02 1.9 0.04 
13 0.05 0.13 0.6 0.05 
14 0.05 0.11 0.05 
15 0.05 0.00 3.8 0.05 
16 0.06 0.72 0.06 
17 0.08 0.27 11.4 0.08 
18 0.09 0.00 0.4 0.09 
19 0.10 0.47 0.7 0.10 
20 0.10 0.00 1.5 0.10 
21 0.12 0.46 0.2 0.12 
22 0.14 0.06 9.8 0.14 
23 0.14 0.01 26.9 0.14 
24 0.15 3.5 10.64 12 0.15 
25 0.44 1.50 2.2 0.44 
26 0.52 0.07 2.2 0.52 
27 0.54 1.07 0.1 0.54 
28 0.69 0.09 0.69 
29 1.04 0.16 1.04 
30 1.71 0.05 6.8 1.71 
31 3.08 0.40 3.1 3.08 
32 0.9 4.12 1.65 0.4 4.12 
33 4.29 0.74 4.1 4.29 
34 4.41 1.37 4.41 
35 1.4 4.50 2.2 0.00 8.9 4.50 
36 4.77 4.13 0.4 4.77 
37 9.00 2.28 9.00 
38 n.e. 9.72 n.e. 0.11 n.e. 9.72 
39 3.6 11.23 6.13 2.2 11.23 

NOTE: IHC: D, decreased expression; P, preserved expression; WB, number means expression value. 0, undetectable; n.e., not examined. qPCR: number means expression value. p14ARF, p16INK4a mRNA: CDKN2A mRNA; p15INK4b mRNA: CDKN2B mRNA.

mRNA expression assay by quantitative RT-PCR

The expression levels of CDKN2A mRNA (0.00–11.23, median 0.10) were significantly associated with both p14ARF and p16INK4a immunohistochemical protein expressions (Table 2, Fig. 2A and B). The expression levels of CDKN2B mRNA (0.00–10.64, median 0.095) were also correlated to p15INK4b protein expressions (Table 2, Fig. 2C).

Figure 2.

Association between the expression levels of CDKN2A mRNA and p14ARF protein (A), CDKN2A mRNA and p16INK4a protein (B), and CDKN2B mRNA and p15INK4b protein (C). There were significant associations between the mRNA and protein expressions of p14ARF, p15INK4b, and p16INK4a, respectively, with P values shown at the top right of the figures. D, association between mRNA levels and gene alterations of CDKN2A. The gene alterations were significantly correlated to the low levels of CDKN2A mRNA (P = 0.0046).

Figure 2.

Association between the expression levels of CDKN2A mRNA and p14ARF protein (A), CDKN2A mRNA and p16INK4a protein (B), and CDKN2B mRNA and p15INK4b protein (C). There were significant associations between the mRNA and protein expressions of p14ARF, p15INK4b, and p16INK4a, respectively, with P values shown at the top right of the figures. D, association between mRNA levels and gene alterations of CDKN2A. The gene alterations were significantly correlated to the low levels of CDKN2A mRNA (P = 0.0046).

Close modal

Gene alteration assessment by multiplex real-time PCR, PCR-SSCP, and MSP

Homozygous deletion (HD) of the CDKN2A gene was detected in 9 of 27 cases (33.3%) by multiplex real-time PCR assay. The remaining 18 cases without homozygous deletion were also analyzed by PCR-SSCP and MSP. No mutation was found in exons of CDKN2A by PCR-SSCP and the subsequent DNA sequencing (data not shown). MSP revealed that no methylation occurred in either the p14ARF or p16INK4a promoter region (data not shown). CDKN2A gene alterations represented by HD were significantly associated with low CDKN2A mRNA levels (Fig. 2D). All but one of the cases with HD in the CDKN2A gene showed decreased immunoreactivity for both p14ARF and p16INK4a.

Survival analysis

Univariate prognostic analysis revealed that decreased expressions of p14ARF and p16INK4a were significantly associated with decreased probability of overall survival, respectively (Table 3, Fig. 3A and B). Patients with decreased expression of p15INK4b showed a strong tendency to have poor prognosis (P = 0.051; Table 3, Fig. 3C). Each of the following was associated with poor prognosis: tumor location in the trunk, tumor location in deep tissue, large tumor size (more than 5 cm), high Ki-67–labeling index (more than 30%), high histological grade (grade 2 or more), and advanced AJCC stage (stage II or higher).

Figure 3.

The Kaplan–Meier curves for overall survival according to p14ARF (A), p16INK4a (B), and p15INK4b (C) expressions. Decreased expressions of p14ARF and p16INK4a were associated with a decreased probability of overall survival, respectively, with P values shown at the top right of the figures. Decreased expression of p15INK4b showed a strong tendency toward association with poor prognosis (P = 0.051). D, the Kaplan–Meier curves for overall survival according to the number of inactivated genes among p14ARF, p15INK4b, and p16INK4a. Patients with inactivation of 2 or more genes showed worse prognosis than those with 1 or no gene inactivated (P < 0.001). E, the Kaplan–Meier curves for overall survival in patients with p16INK4a inactivation according to the p15INK4b expression. Patients with preserved expression of p15INb4b in the absence of p16INK4a show better prognosis than the p15INK4b and p16INK4a coinactivated cases (P = 0.042).

Figure 3.

The Kaplan–Meier curves for overall survival according to p14ARF (A), p16INK4a (B), and p15INK4b (C) expressions. Decreased expressions of p14ARF and p16INK4a were associated with a decreased probability of overall survival, respectively, with P values shown at the top right of the figures. Decreased expression of p15INK4b showed a strong tendency toward association with poor prognosis (P = 0.051). D, the Kaplan–Meier curves for overall survival according to the number of inactivated genes among p14ARF, p15INK4b, and p16INK4a. Patients with inactivation of 2 or more genes showed worse prognosis than those with 1 or no gene inactivated (P < 0.001). E, the Kaplan–Meier curves for overall survival in patients with p16INK4a inactivation according to the p15INK4b expression. Patients with preserved expression of p15INb4b in the absence of p16INK4a show better prognosis than the p15INK4b and p16INK4a coinactivated cases (P = 0.042).

Close modal
Table 3.

Univariate analysis for overall survival

VariableNo. of patients5-y survial rateP
Age, y    
 <50 45 51.4 0.41 
 ≥50 37 53.2  
Sex 
 Male 43 47.4 0.28 
 Female 39 57.4  
NF1 
 Present 32 41.1 0.26 
 Absent 50 57.7  
Site 
 Extremity 36 64.3 0.041 
 Trunk 46 42.5  
Tumor depth 
 Superficial 20 78.6 0.004 
 Deep 62 43.4  
Tumor size 
 <5cm 23 78.3 0.006 
 ≥5cm 59 41.7  
Adjuvant therapy 
 Given 50.0 0.47 
 Not given 79 52.0  
Tumor necrosis 
 Absent 38 63.4 0.067 
 Present 44 42.3  
Mitotic counts 
 0–9/10HPF 56 53.7 0.43 
 ≥10/10HPF 26 47.1  
Ki-67–labeling index    
 0%–29% 58 63.8 <0.001 
 ≥30% 24 21.0  
Microvessel density    
 <15/HPF 39 65.6 0.06 
 ≥15/HPF 43 39.7  
Rhabdomyoblastic differentiation 
 Present 10 33.8 0.20 
 Absent 72 54.5  
Epithelioid feature 
 Present 20.0 0.11 
 Absent 77 54.6  
FNCLCC grading 
 1 25 72.5 0.030 
 2 + 3 57 43.4  
AJCC staging 
 I 25 72.5 0.030 
 II + III + IV 57 43.4  
p14ARF IHC 
 Preserved 42 74.1 <0.001 
 Decreased 40 28.5  
p15INK4b IHC 
 Preserved 38 67.9 0.051 
 Decreased 44 38.9  
p16INK4a IHC 
 Preserved 41 68.9 0.002 
 Decreased 41 34.0  
VariableNo. of patients5-y survial rateP
Age, y    
 <50 45 51.4 0.41 
 ≥50 37 53.2  
Sex 
 Male 43 47.4 0.28 
 Female 39 57.4  
NF1 
 Present 32 41.1 0.26 
 Absent 50 57.7  
Site 
 Extremity 36 64.3 0.041 
 Trunk 46 42.5  
Tumor depth 
 Superficial 20 78.6 0.004 
 Deep 62 43.4  
Tumor size 
 <5cm 23 78.3 0.006 
 ≥5cm 59 41.7  
Adjuvant therapy 
 Given 50.0 0.47 
 Not given 79 52.0  
Tumor necrosis 
 Absent 38 63.4 0.067 
 Present 44 42.3  
Mitotic counts 
 0–9/10HPF 56 53.7 0.43 
 ≥10/10HPF 26 47.1  
Ki-67–labeling index    
 0%–29% 58 63.8 <0.001 
 ≥30% 24 21.0  
Microvessel density    
 <15/HPF 39 65.6 0.06 
 ≥15/HPF 43 39.7  
Rhabdomyoblastic differentiation 
 Present 10 33.8 0.20 
 Absent 72 54.5  
Epithelioid feature 
 Present 20.0 0.11 
 Absent 77 54.6  
FNCLCC grading 
 1 25 72.5 0.030 
 2 + 3 57 43.4  
AJCC staging 
 I 25 72.5 0.030 
 II + III + IV 57 43.4  
p14ARF IHC 
 Preserved 42 74.1 <0.001 
 Decreased 40 28.5  
p15INK4b IHC 
 Preserved 38 67.9 0.051 
 Decreased 44 38.9  
p16INK4a IHC 
 Preserved 41 68.9 0.002 
 Decreased 41 34.0  

A multivariate analysis carried out using variables that were related to poor prognosis in the univariate analysis revealed that tumor location in the trunk, tumor location in deep tissue, and decreased expressions of either p14ARF or p16INK4a persisted as independent risk factors for a poor outcome (Table 4).

Table 4.

Multivariate analysis for overall survival

VariableHazard ratio95% CIP
Site 
 Extremity   
 Trunk 2.08 1.44–2.99 0.042 
Tumor depth 
 Superficial   
 Deep 3.93 2.05–7.56 0.025 
Tumor size 
 <5cm   
 ≥5cm 1.26 0.71–2.25 0.69 
Ki-67–labeling index 
 0%–29%   
 ≥30% 1.36 0.89–2.08 0.47 
FNCLCC grading 
 1   
 2 + 3 1.83 1.15–2.92 1.00 
AJCC staging 
 I   
 II + III + IV 1–1 1.00 
p14ARF IHC 
 Preserved   
 Decreased 2.72 1.75–4.21 0.021 
p16INK4a IHC 
 Preserved   
 Decreased 2.21 1.52–3.22 0.034 
VariableHazard ratio95% CIP
Site 
 Extremity   
 Trunk 2.08 1.44–2.99 0.042 
Tumor depth 
 Superficial   
 Deep 3.93 2.05–7.56 0.025 
Tumor size 
 <5cm   
 ≥5cm 1.26 0.71–2.25 0.69 
Ki-67–labeling index 
 0%–29%   
 ≥30% 1.36 0.89–2.08 0.47 
FNCLCC grading 
 1   
 2 + 3 1.83 1.15–2.92 1.00 
AJCC staging 
 I   
 II + III + IV 1–1 1.00 
p14ARF IHC 
 Preserved   
 Decreased 2.72 1.75–4.21 0.021 
p16INK4a IHC 
 Preserved   
 Decreased 2.21 1.52–3.22 0.034 

In terms of p14ARF and p16INK4a expressions in the primary tumors, decreased expression of both p14ARF and p16INK4a was observed in 28 of the 82 patients (34.1%), whereas preserved expression of both was detected in 29 patients (35.4%; Table 5). A combined prognostic evaluation with grouping by p14ARF and p16INK4a expressions revealed that the 5-year overall survival rate of cases with a decrease in the expression of both proteins was 25.8%, whereas that of cases in which the expression of both proteins preserved was 83.3%, with the difference in the 2 values being statistically significant (P < 0.001).

Table 5.

Combinational prognostic analysis of p14ARF, p15INK4b, and p16INK4a expressions

VariablesNo. of patients5-y survial rateP
Combinational prognostic analysis of p14ARF and p16INK4a expressions p14ARF p16INK4a    
 28 (34.1%) 25.8 <0.001 (DD vs. PP) 
 12 (14.6%) 31.3  
 13 (15.9%) 51.1  
 29 (35.4%) 83.3  
The number of inactivated genes among p14ARF, p15INK4b, and p16INK4a  20 (24.4%) 77.1 <0.001 (0 + 1 vs. 2 + 3) 
  22 (26.8%) 71.4  
  17 (20.7%) 38.2  
  23 (28.0%) 21.6  
Prognostic effect of p15INK4b expression in the absence of p16INK4a p15INK4b p16INK4a    
 30 (73.2%) 24.8 0.042 
 11 (26.8%) 75  
VariablesNo. of patients5-y survial rateP
Combinational prognostic analysis of p14ARF and p16INK4a expressions p14ARF p16INK4a    
 28 (34.1%) 25.8 <0.001 (DD vs. PP) 
 12 (14.6%) 31.3  
 13 (15.9%) 51.1  
 29 (35.4%) 83.3  
The number of inactivated genes among p14ARF, p15INK4b, and p16INK4a  20 (24.4%) 77.1 <0.001 (0 + 1 vs. 2 + 3) 
  22 (26.8%) 71.4  
  17 (20.7%) 38.2  
  23 (28.0%) 21.6  
Prognostic effect of p15INK4b expression in the absence of p16INK4a p15INK4b p16INK4a    
 30 (73.2%) 24.8 0.042 
 11 (26.8%) 75  

NOTE: D, decreased; P, preserved; DD, decreased in both p14ARF and p16INK4a; PP, preserved in both p14ARF and p16INK4a.

There was an association between the prognosis and the number of inactivations among p14ARF, p15INK4b, and p16INK4a; the cases with 2 or 3 inactivated genes showed a significantly more adverse prognosis than those with only 1 or no inactivated genes (P < 0.001; Table 5, Fig. 3D).

Finally, the prognostic significance of p15INK4b expression in the absence of p16INK4a was evaluated. Among the 41 cases with p16INK4a inactivation, 30 cases (73.2%) showed decreased expression of p15INK4b, and the remaining 11 cases (26.8%) showed preserved expression of p15INK4b (Table 5). The 5-year overall survival rate of patients with coinactivation of p15INK4b and p16INK4a was 24.8%, whereas that of patients with preserved p15INK4b was 75.0%, which was significantly higher than that of coinactivated patients (Fig. 3E).

Our univariate prognostic analysis of p14ARF, p15INK4b, and p16INK4a inactivation in MPNSTs revealed that each gene inactivation was significantly or almost significantly associated with poor prognosis. Furthermore, the prognostic importance of p14ARF and p16INK4a status in MPNSTs was reconfirmed by the multivariate analysis. A combined prognostic analysis showed that MPNSTs with inactivation of 2 or more genes among p14ARF, p15INK4b, and p16INK4a showed much worse prognosis than those with inactivation of 1 or no genes. The above results indicate that a synergistic effect of the combined deficiency for p14ARF, p15INK4b, and p16INK4a induced a further high-grade malignancy in MPNSTs. The results from this study contribute not only to estimation of the molecular pathophysiology of MPNST, but also to the improvement of clinical management of patients with MPNST through the provision of precise prognostic prediction.

The impact of p15INK4b inactivation in human cancers has been less clear than that of the p14ARF and p16INK4a inactivations (15). But recently, the significance of p15INK4b inactivation in malignant tumors has got a lot of attention. The loss of CDKN2B encoding p15INK4b is observed in various types of malignant tumors, but the frequent loss of CDKN2B in human tumors could be explained by its juxtaposition to CDKN2A, in which proximity could lead to frequent codeletion. A recent study using a mouse model has proposed the interesting hypothesis that p15INK4b functions as a critical tumor suppressor in the absence of p16INK4a (15). In human tumors, the tumor-suppressive effect of p15INK4b in the setting of p16INK4a inactivation has not been investigated well. This is the first report evaluating the prognostic role of p15INK4b in the absence of p16INK4a in MPNSTs. Our prognostic analysis of p16INK4a-inactivated cases showed that p15INK4b-preserved cases showed significantly better prognosis than p15INK4b-inactivated cases. This finding indirectly supports the hypothesis that p15INK4b provides a critical backup function for p16INK4a in MPNSTs. In addition, it may provide an explanation for the frequent loss of the complete CDKN2BCDKN2A locus in human high-grade malignancies, including MPNST (15, 43).

Here we revealed the frequency of inactivation of p14ARF, p15INK4b, and p16INK4a in MPNSTs by a large-scale clinicopathological study with multiple methods. Inactivation of p14ARF, p15INK4b, and p16INK4a was observed in 50%, 49%, and 53% of MPNSTs, respectively. In previous studies, the frequency of p14ARF, p15INK4b, and p16INK4a inactivation in MPNSTs has been a controversial issue. Some papers have stated that p16INK4a inactivation in neurofibroma is a critical process in progression to MPNST, and p16INK4a is inactivated in almost all MPNSTs (18, 19, 44). On the other hand, some recent papers have indicated that 9p21-related genes including p16INK4a are inactivated in only half of all cases of MPNST (24, 43, 45). Our data support the latter statement: we found that inactivation of the p14ARF, p15INK4b, and p16INK4a genes was involved in high-grade malignant transformation in about half of the MPNSTs studied.

CDKN2A is known to exhibit more HDs than other recessive cancer genes in various types of cancers (46). This may be because it is adjacent to a large gene desert and therefore HDs are associated with less negative selection (46). Our study showed that HD of CDKN2A was detected in 33.3% of the cases, whereas neither point mutation of CDKN2A nor methylation of the p14ARF and p16INK4a promoter regions was detected. In addition to our study, several other authors have investigated gene alterations of CDKN2A in MPNSTs (16–23, 42–44, 47). According to their reports, HD is the predominant cause of inactivation of the 9p21 locus in MPNSTs, and the rate of HD ranges from 7.1% to 56.5% (16–20, 22, 43, 44, 47). Point mutation of CDKN2A has been reported in only 1 MPNST case (17). Methylation of the p14ARF and p16INK4a promoter regions has been reported in only 2 cases (7.7% of the investigated cases), respectively (43). Our results are basically consistent with those of the previous reports, and indicate that the main alteration causing CDKN2A gene inactivation is HD rather than point mutation or methylation of the promoter regions in MPNSTs.

NF1-related MPNST has an NF1 gene dysfunction leading to an excessive activation of the Ras signal pathway, whereas sporadic MPNST does not always show an NF1 gene dysfunction (48). The pathogenesis of MPNST is a fascinating subject, because both NF1-related and sporadic MPNSTs show similar histological findings and molecular characteristics despite their difference in genetic background. Our study showed that there was no significant difference in the expressions of p14ARF, p15INK4b, and p16INK4a between NF1-related and sporadic MPNSTs. Perrone and colleagues reported that inactivation of 1 or more 9p21-related genes detected by RT-PCR was found in 11 of 14 NF1-related (78%) and 9 of 12 sporadic (75%) cases, respectively (43). They concluded that the inactivation profile of 9p21-related genes showed no significant difference between NF1-related and sporadic MPNSTs. Our findings were basically consistent with those of this previous report.

In summary, we carried out a comprehensive analysis of p14ARF, p15INK4b, and p16INK4a expressions in MPNSTs. Inactivation of p14ARF, p15INK4b, and p16INK4a was associated with poor prognosis, respectively, and a combined inactivation of them leaded to worse prognosis in MPNSTs. We conclude that a combined evaluation of p14ARF, p15INK4b, and p16INK4a status provides useful prognostic information for patients with MPNSTs.

The authors appreciate for the technical supports from Ms. Tateishi and the Research Support Center, Graduate School of Medical Sciences, Kyushu University. They also appreciate KN International for revising the English used in this article.

Y. Oda is supported by a Grant-in-Aid for Scientific Research (B; grant no. 21390107) from the Japan Society for the Promotion of Science, Tokyo, Japan.

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.
Anghileri
M
,
Miceli
R
,
Fiore
M
,
Mariani
L
,
Ferrari
A
,
Mussi
C
, et al
Malignant peripheral nerve sheath tumors: prognostic factors and survival in a series of patients treated at a single institution
.
Cancer
2006
;
107
:
1065
74
.
2.
Ducatman
BS
,
Scheithauer
BW
,
Piepgras
DG
,
Reiman
HM
,
Ilstrup
DM
. 
Malignant peripheral nerve sheath tumors. A clinicopathologic study of 120 cases
.
Cancer
1986
;
57
:
2006
21
.
3.
Cichowski
K
,
Shih
TS
,
Schmitt
E
,
Santiago
S
,
Reilly
K
,
McLaughlin
ME
, et al
Mouse models of tumor development in neurofibromatosis type 1
.
Science
1999
;
286
:
2172
6
.
4.
Vogel
KS
,
Klesse
LJ
,
Velasco-Miguel
S
,
Meyers
K
,
Rushing
EJ
,
Parada
LF
. 
Mouse tumor model for neurofibromatosis type 1
.
Science
1999
;
286
:
2176
9
.
5.
Hummel
TR
,
Jessen
W
,
Miller
SC
,
Kluwe
L
,
Mautner
V
,
Wallace
MR
, et al
Gene expression analysis identifies potential biomarkers of neurofibromatosis type 1 including adrenomedullin
.
Clin Cancer Res
. 
2010
;
16
:
5048
57
.
6.
Levy
P
,
Ripoche
H
,
Laurendeau
I
,
Lazar
V
,
Ortonne
N
,
Parfait
B
, et al
Microarray-based identification of tenascin C and tenascin XB, genes possibly involved in tumorigenesis associated with neurofibromatosis type 1
.
Clin Cancer Res
2007
;
13
:
398
407
.
7.
Sordillo
PP
,
Helson
L
,
Hajdu
SI
,
Magill
GB
,
Kosloff
C
,
Golbey
RB
, et al
Malignant schwannoma—clinical characteristics, survival, and response to therapy
.
Cancer
1981
;
47
:
2503
9
.
8.
Hruban
RH
,
Shiu
MH
,
Senie
RT
,
Woodruff
JM
. 
Malignant peripheral nerve sheath tumors of the buttock and lower extremity. A study of 43 cases
.
Cancer
1990
;
66
:
1253
65
.
9.
Kawaguchi
K
,
Oda
Y
,
Saito
T
,
Yamamoto
H
,
Tamiya
S
,
Takahira
T
, et al
Mechanisms of inactivation of the p16INK4a gene in leiomyosarcoma of soft tissue: decreased p16 expression correlates with promoter methylation and poor prognosis
.
J Pathol
2003
;
201
:
487
95
.
10.
Oda
Y
,
Yamamoto
H
,
Takahira
T
,
Kobayashi
C
,
Kawaguchi
K
,
Tateishi
N
, et al
Frequent alteration of p16(INK4a)/p14(ARF) and p53 pathways in the round cell component of myxoid/round cell liposarcoma: p53 gene alterations and reduced p14(ARF) expression both correlate with poor prognosis
.
J Pathol
2005
;
207
:
410
21
.
11.
Perrone
F
,
Tamborini
E
,
Dagrada
GP
,
Colombo
F
,
Bonadiman
L
,
Albertini
V
, et al
9p21 locus analysis in high-risk gastrointestinal stromal tumors characterized for c-kit and platelet-derived growth factor receptor alpha gene alterations
.
Cancer
2005
;
104
:
159
69
.
12.
Cao
J
,
Zhou
J
,
Gao
Y
,
Gu
L
,
Meng
H
,
Liu
H
, et al
Methylation of p16 CpG island associated with malignant progression of oral epithelial dysplasia: a prospective cohort study
.
Clin Cancer Res
2009
;
15
:
5178
83
.
13.
Hallor
KH
,
Staaf
J
,
Bovee
JV
,
Hogendoorn
PC
,
Cleton-Jansen
AM
,
Knuutila
S
, et al
Genomic profiling of chondrosarcoma: chromosomal patterns in central and peripheral tumors
.
Clin Cancer Res
2009
;
15
:
2685
94
.
14.
Latres
E
,
Malumbres
M
,
Sotillo
R
,
Martin
J
,
Ortega
S
,
Martin-Caballero
J
, et al
Limited overlapping roles of P15(INK4b) and P18(INK4c) cell cycle inhibitors in proliferation and tumorigenesis
.
EMBO J
2000
;
19
:
3496
506
.
15.
Krimpenfort
P
,
Ijpenberg
A
,
Song
JY
,
Van Der Valk
M
,
Nawijn
M
,
Zevenhoven
J
, et al
p15Ink4b is a critical tumour suppressor in the absence of p16Ink4a
.
Nature
2007
;
448
:
943
6
.
16.
Berner
JM
,
Sorlie
T
,
Mertens
F
,
Henriksen
J
,
Saeter
G
,
Mandahl
N
, et al
Chromosome band 9p21 is frequently altered in malignant peripheral nerve sheath tumors: studies of CDKN2A and other genes of the pRB pathway
.
Genes Chromosomes Cancer
1999
;
26
:
151
60
.
17.
Kourea
HP
,
Orlow
I
,
Scheithauer
BW
,
Cordon-Cardo
C
,
Woodruff
JM
. 
Deletions of the INK4A gene occur in malignant peripheral nerve sheath tumors but not in neurofibromas
.
Am J Pathol
1999
;
155
:
1855
60
.
18.
Nielsen
GP
,
Stemmer-Rachamimov
AO
,
Ino
Y
,
Moller
MB
,
Rosenberg
AE
,
Louis
DN
. 
Malignant transformation of neurofibromas in neurofibromatosis 1 is associated with CDKN2A/p16 inactivation
.
Am J Pathol
1999
;
155
:
1879
84
.
19.
Birindelli
S
,
Perrone
F
,
Oggionni
M
,
Lavarino
C
,
Pasini
B
,
Vergani
B
, et al
Rb and TP53 pathway alterations in sporadic and NF1-related malignant peripheral nerve sheath tumors
.
Lab Invest
2001
;
81
:
833
44
.
20.
Perry
A
,
Kunz
SN
,
Fuller
CE
,
Banerjee
R
,
Marley
EF
,
Liapis
H
, et al
Differential NF1, p16, and EGFR patterns by interphase cytogenetics (FISH) in malignant peripheral nerve sheath tumor (MPNST) and morphologically similar spindle cell neoplasms
.
J Neuropathol Exp Neurol
2002
;
61
:
702
9
.
21.
Upadhyaya
M
,
Han
S
,
Consoli
C
,
Majounie
E
,
Horan
M
,
Thomas
NS
, et al
Characterization of the somatic mutational spectrum of the neurofibromatosis type 1 (NF1) gene in neurofibromatosis patients with benign and malignant tumors
.
Hum Mutat
2004
;
23
:
134
46
.
22.
Holtkamp
N
,
Malzer
E
,
Zietsch
J
,
Okuducu
AF
,
Mucha
J
,
Mawrin
C
, et al
EGFR and erbB2 in malignant peripheral nerve sheath tumors and implications for targeted therapy
.
Neuro Oncol
2008
;
10
:
946
57
.
23.
Spurlock
G
,
Knight
SJ
,
Thomas
N
,
Kiehl
TR
,
Guha
A
,
Upadhyaya
M
. 
Molecular evolution of a neurofibroma to malignant peripheral nerve sheath tumor (MPNST) in an NF1 patient: correlation between histopathological, clinical and molecular findings
.
J Cancer Res Clin Oncol
2010
;
136
:
1869
80
.
24.
Mantripragada
KK
,
Spurlock
G
,
Kluwe
L
,
Chuzhanova
N
,
Ferner
RE
,
Frayling
IM
, et al
High-resolution DNA copy number profiling of malignant peripheral nerve sheath tumors using targeted microarray-based comparative genomic hybridization
.
Clin Cancer Res
2008
;
14
:
1015
24
.
25.
Scheithauer
BW
,
Louis
DN
,
Hunter
S
,
Woodruff
JM
,
Antonescu
CR
. 
Malignant peripheral nerve sheath tumour (MPNST)
.
In:
Louis
DN
,
Ohgaki
H
,
Wiestler
OD
,
editors
. 
WHO Classification of Tumours of the Central Nervous System
.
Lyon, France
:
IARC
; 
2007
.
p. 160
2
.
26.
Malignant tumors of the peripheral nerves
.
In:
Weiss
SW
,
Goldblum
JR
,
editors
. 
Enzinger and Weiss's Soft Tissue Tumors
. 5th ed.
St. Louis, MO
:
Mosby
; 
2008
.
p. 903
44
.
27.
Neurofibromatosis
. 
Conference statement. National Institutes of Health Consensus Development Conference
.
Arch Neurol
1988
;
45
:
575
8
.
28.
Guillou
L
,
Coindre
JM
,
Bonichon
F
,
Nguyen
BB
,
Terrier
P
,
Collin
F
, et al
Comparative study of the National Cancer Institute and French Federation of Cancer Centers Sarcoma Group grading systems in a population of 410 adult patients with soft tissue sarcoma
.
J Clin Oncol
1997
;
15
:
350
62
.
29.
Edge
SB
,
Byrd
DR
,
Compton
CC
,
Fritz
AG
,
Greene
FL
,
Trotti
AIII
editors
. 
AJCC Cancer Staging Manual
. 
7th ed
.
New York
:
Springer
; 
2010
.
30.
Iwasa
A
,
Oda
Y
,
Kurihara
S
,
Ohishi
Y
,
Yasunaga
M
,
Nishimura
I
, et al
Malignant transformation of mature cystic teratoma to squamous cell carcinoma involves altered expression of p53- and p16/Rb-dependent cell cycle regulator proteins
.
Pathol Int
2008
;
58
:
757
64
.
31.
Child
FJ
,
Scarisbrick
JJ
,
Calonje
E
,
Orchard
G
,
Russell-Jones
R
,
Whittaker
SJ
. 
Inactivation of tumor suppressor genes p15(INK4b) and p16(INK4a) in primary cutaneous B cell lymphoma
.
J Invest Dermatol
2002
;
118
:
941
8
.
32.
Kobayashi
C
,
Oda
Y
,
Takahira
T
,
Izumi
T
,
Kawaguchi
K
,
Yamamoto
H
, et al
Chromosomal aberrations and microsatellite instability of malignant peripheral nerve sheath tumors: a study of 10 tumors from nine patients
.
Cancer Genet Cytogenet
2006
;
165
:
98
105
.
33.
Hemmerlein
B
,
Galuschka
L
,
Putzer
N
,
Zischkau
S
,
Heuser
M
. 
Comparative analysis of COX-2, vascular endothelial growth factor and microvessel density in human renal cell carcinomas
.
Histopathology
2004
;
45
:
603
11
.
34.
Kohashi
K
,
Oda
Y
,
Yamamoto
H
,
Tamiya
S
,
Matono
H
,
Iwamoto
Y
, et al
Reduced expression of SMARCB1/INI1 protein in synovial sarcoma
.
Mod Pathol
23
;
981
90
.
35.
Kobayashi
C
,
Oda
Y
,
Takahira
T
,
Izumi
T
,
Kawaguchi
K
,
Yamamoto
H
, et al
Aberrant expression of CHFR in malignant peripheral nerve sheath tumors
.
Mod Pathol
2006
;
19
:
524
32
.
36.
Nagashima
Y
,
Ohaki
Y
,
Tanaka
Y
,
Sumino
K
,
Funabiki
T
,
Okuyama
T
, et al
Establishment of an epithelioid malignant schwannoma cell line (YST-1)
.
Virchows Arch B Cell Pathol Incl Mol Pathol
1990
;
59
:
321
7
.
37.
Matsuura
S
,
Oda
Y
,
Matono
H
,
Izumi
T
,
Yamamoto
H
,
Tamiya
S
, et al
Overexpression of A disintegrin and metalloproteinase 28 is correlated with high histologic grade in conventional chondrosarcoma
.
Hum Pathol
.
41
:
343
51
.
38.
Kohashi
K
,
Oda
Y
,
Yamamoto
H
,
Tamiya
S
,
Oshiro
Y
,
Izumi
T
, et al
SMARCB1/INI1 protein expression in round cell soft tissue sarcomas associated with chromosomal translocations involving EWS: a special reference to SMARCB1/INI1 negative variant extraskeletal myxoid chondrosarcoma
.
Am J Surg Pathol
2008
;
32
:
1168
74
.
39.
Carter
TL
,
Reaman
GH
,
Kees
UR
. 
INK4A/ARF deletions are acquired at relapse in childhood acute lymphoblastic leukaemia: a paired study on 25 patients using real-time polymerase chain reaction
.
Br J Haematol
2001
;
113
:
323
8
.
40.
Berggren
P
,
Kumar
R
,
Sakano
S
,
Hemminki
L
,
Wada
T
,
Steineck
G
, et al
Detecting homozygous deletions in the CDKN2A(p16(INK4a))/ARF(p14(ARF)) gene in urinary bladder cancer using real-time quantitative PCR
.
Clin Cancer Res
2003
;
9
:
235
42
.
41.
Saito
T
,
Oda
Y
,
Sakamoto
A
,
Tamiya
S
,
Kinukawa
N
,
Hayashi
K
, et al
Prognostic value of the preserved expression of the E-cadherin and catenin families of adhesion molecules and of beta-catenin mutations in synovial sarcoma
.
J Pathol
2000
;
192
:
342
50
.
42.
Kawaguchi
K
,
Oda
Y
,
Saito
T
,
Yamamoto
H
,
Takahira
T
,
Kobayashi
C
, et al
DNA hypermethylation status of multiple genes in soft tissue sarcomas
.
Mod Pathol
2006
;
19
:
106
14
.
43.
Perrone
F
,
Tabano
S
,
Colombo
F
,
Dagrada
G
,
Birindelli
S
,
Gronchi
A
, et al
p15INK4b, p14ARF, and p16INK4a inactivation in sporadic and neurofibromatosis type 1-related malignant peripheral nerve sheath tumors
.
Clin Cancer Res
2003
;
9
:
4132
8
.
44.
Agesen
TH
,
Florenes
VA
,
Molenaar
WM
,
Lind
GE
,
Berner
JM
,
Plaat
BE
, et al
Expression patterns of cell cycle components in sporadic and neurofibromatosis type 1-related malignant peripheral nerve sheath tumors
.
J Neuropathol Exp Neurol
2005
;
64
:
74
81
.
45.
Zhou
H
,
Coffin
CM
,
Perkins
SL
,
Tripp
SR
,
Liew
M
,
Viskochil
DH
. 
Malignant peripheral nerve sheath tumor: a comparison of grade, immunophenotype, and cell cycle/growth activation marker expression in sporadic and neurofibromatosis 1-related lesions
.
Am J Surg Pathol
2003
;
27
:
1337
45
.
46.
Bignell
GR
,
Greenman
CD
,
Davies
H
,
Butler
AP
,
Edkins
S
,
Andrews
JM
, et al
Signatures of mutation and selection in the cancer genome
.
Nature
463
:
893
8
.
47.
Orlow
I
,
Drobnjak
M
,
Zhang
ZF
,
Lewis
J
,
Woodruff
JM
,
Brennan
MF
, et al
Alterations of INK4A and INK4B genes in adult soft tissue sarcomas: effect on survival
.
J Natl Cancer Inst
1999
;
91
:
73
9
.
48.
Grobmyer
SR
,
Reith
JD
,
Shahlaee
A
,
Bush
CH
,
Hochwald
SN
. 
Malignant Peripheral Nerve Sheath Tumor: molecular pathogenesis and current management considerations
.
J Surg Oncol
2008
;
97
:
340
9
.