Skip to main content
  • Review
  • Published:

The CDKN2A (p16) Gene and Human Cancer

Summary

CDKN2A, the gene encoding the cell-cycle inhibitor p16CDKN2A, was first identified in 1994. Since then, somatic mutations have been observed in many cancers and germline alterations have been found in kindreds with familial atypical multiple mole/melanoma (FAMMM), also known as atypical mole syndrome. In this review we tabulate the known mutations in this gene and discuss specific aspects, particularly with respect to germline mutations and cancer predisposition.

Introduction

CDKN2A—the Gene

CDKN2A has been given different names (p16INK4, p16INK4A, CDK4I, MTS1, and p16) by different investigators, but was recently assigned the designation CDKN2A (for cyclin dependent kinase inhibitor 2A) by the Human Genome Organisation nomenclature committee. The gene is composed of 3 exons, with one alternatively spliced exon (E1-β). It is situated on chromosome 9p21, in a region that shows a high frequency of loss of heterozygosity (LOH) in numerous tumor types (1). The gene itself is mutated (or inactivated in some way) in many types of human cancers. In this sense, CDKN2A bears a striking resemblance to the paradigmatic tumor suppressor gene, p53. CDKN2A may prove to be as important a regulator of cell growth as p53.

An interesting parallel between these two genes can be drawn upon examination of the tumor spectrum observed in families carrying germline mutations. Although both genes are somatically altered in a wide variety of cancers, there is a more specific pattern of malignancy associated with germline mutations. For instance, CDKN2A somatic mutations (mainly deletions) are common in glioblastoma (2), but a case of glioblastoma has not yet been reported in a familial atypical multiple mole/melanoma (FAMMM) kindred, where cutaneous malignant melanoma is by far the commonest cancer (3). Similarly, colorectal and ovarian cancer frequently exhibit somatic p53 mutations (4). However, these cancers are rarely seen as part of the familial Li-Fraumeni syndrome, which typically features soft tissue sarcomas, leukemia, and brain cancers in children, and breast cancer in young women (5).

p16CDK2V2A—the Protein

CDKN2A encodes a 156 amino acid, 16kD cell-cycle inhibitor protein, which normally blocks abnormal cell growth and proliferation by binding to complexes of cyclin-dependent kinases (CDK) 4 and 6, and cyclin D. This binding inhibits the kinase activity of the enzyme, which arrests the cell cycle in the Gl phase (Fig. 1). Mutant p16CDKN2A is unable to form stable complexes with the enzyme and, therefore, does not effectively inhibit progression of cells through inappropriate mitotic divisions. The importance of this protein as a cell-cycle regulator is demonstrated by the wide array of tumor types in which mutations of CDKN2A have been observed.

Fig. 1
figure 1

Diagrammatic representation of the involvement of (p16) in cell cycle regulation

Abbreviations are as follows: CDK, cyclin-dependent kinase; PCNA, proliferating cell nuclear antigen; Rb, retinoblastoma gene product; E2F, transcription factor; P, phosphate; Tx genes, gene transcription; p16, CDKN2A gene product.

Identification of CDKN2A as a Possible Tumor Suppressor Gene

Molecular cytogenetic studies revealed frequent heterozygous and homozygous deletions of the chromosome 9p21-p22 region in melanoma and other cell lines (6,7). Linkage to markers from 9p13-p22 was reported in melanoma kindreds from the United States (8,9), Australia (10), Holland (11), and the United Kingdom (12). CDKN2A mutations were subsequently reported in several families within each cohort. However, there appear to be some families that are linked to 9p21 in which mutations in CDKN2A or the closely related adjacent gene, CDKN2B (p15), have not been found.

Homozygous deletions and intragenic mutations in CDKN2A are often observed in human cell lines. Initially, there was some controversy over the significance of these alterations, since they were detected far less frequently in corresponding primary tumors showing LOH at 9p21 (1, 1315). This discrepancy suggested that another tumor suppressor gene is the target of LOH in the region or simply that CDKN2A is important for the maintenance of viability in cell culture. The issue has been clarified to some extent by two recent discoveries that suggest that LOH at 9p21 may be accounted for by previously undetected mutations in CDKN2A. First, microdeletions (spanning less than 200 kilobases and encompassing CDKN2A) are present; these are, however, only detectable using an array of microsatellites close to the gene and have been confirmed by fluorescent in situ hybridization (FISH) (16). Secondly, CDKN2A can be inactivated by methylation of the CpG island 5′ of the coding region (1719). The involvement of several mechanisms in CDKN2A inactivation has been shown in a comprehensive analysis of 29 primary head and neck squamous cell carcinomas. By immunohistochemistry, 24 of the cancers showed absence of nuclear staining. Of these 24 cancers, 16 had homozygous deletions of CDKN2A, 5 had CpG methylation, 1 had a genomic rearrangement, and another had a frameshift mutation in exon 1 (20).

Germline Mutations

Germline mutations in CDKN2A reported to date are primarily point mutations (Fig. 2). They are observed in FAMMM families but also in cancer-prone kindreds which do not fit the criteria for FAMMM.

Fig. 2
figure 2

Germline mutations and polymorphisms in CDKN2A identified to date (July 1996) and their relative frequencies

Although the CDKN2A germline variants are presented here in melanoma kindreds, this term does not apply to all of the families listed here: the Met53lle (23), Gly101Trp (33), and 12 bp del 268–279 (34) have all been reported to occur in kindreds with only one case of melanoma. Each symbol to the left of the diagram represents one family in which the corresponding mutation has been found. Underlined mutations occur in consensus ankyrin domain amino acids. Mutations in bold are also seen somatically in primary tumors and/or cell lines. Mutations accompanied by asterisks have some kindred sharing a common founder. #In the original reference, this mutation was reported as Ins111Arg. This is probably the same mutation as the Ins113Arg reported by Borg et al. (63).

Familial Atypical Multiple Mole/Melanoma (FAMMM)

Germline mutations in CDKN2A have been reported in FAMMM pedigrees that show linkage to chromosome 9p. Hussussian et al. described eight CDKN2A germline substitutions observed in 13 of 18 American FAMMM kindreds (21). Of these, six are probably disease-related mutations since they were identified in 33 out of 36 melanoma cases in nine families. The remaining two were detected in normal controls and are thought to be nondeleterious polymorphisms.

A 19 base pair germline deletion in CDKN2A was detected in 13 of 15 Dutch FAMMM kindreds (22). This deletion is a founder mutation; the 13 families all originate from the same geographic region and share a common haplotype. CDKN2A mutations were not found in two other Dutch melanoma families. Two individuals from one of the families carrying the 19 bp deletion were homozygous for the mutation. One of these individuals had three mildly atypical nevi and died at the age of 54 from adenocarcinoma originating from an unknown primary site and the other was diagnosed with melanoma in situ at age 15. The fact that these two homozygous individuals did not have more severe phenotypes than their heterozygous relatives suggests that there may be redundancy that compensates for the lack of p16CDKN2A activity.

The Met53Ile mutation (Fig. 2) could also be a founder mutation, since the antecedents of the three Australian melanoma kindreds reported with this mutation all originally lived in Scotland (23) and another report of a family with this mutation identified the place of origin as the west of Scotland (24), although this family does not fit the FAMMM criteria. Haplotype analyses are now underway but have yet to be completed.

CDKN2A, FAMMM, and Pancreatic Cancer

Pancreatic adenocarcinoma is probably the second commonest cancer in FAMMM families. The observed/expected ratio for the frequency of pancreatic cancer among 200 individuals from nine FAMMM families was 13.4 (P < 0.001) (3). In several chromosome 9p-linked FAMMM families, a mutation in CDKN2A was found to cosegregate with both melanoma and pancreatic adenocarcinoma (21,22,25). However, this risk may be limited to a subset of families. In families where the CDKN2A mutation impaired the function of the corresponding protein (p16M) in vitro, the risk of pancreatic cancer was increased 13-fold (standardized incidence ratio: 13.1, 95% CI, 1.5–47.4), whereas no cases of pancreatic cancer were found in families with CDKN2A mutations that did not affect the function of the protein (p16W) in the assay used by this group (25).

The study of Ciotti et al. supports the association of p16M mutations and pancreatic cancer risk in FAMMM families (26). They detected a Gly101Trp missense mutation in seven Italian melanoma-prone kindreds (presumably derived from a common founder) having a combined total of three pancreatic cancers. This mutation affected the function of the p16CDKN2A protein in vitro, and was associated with increased cancer risk (27). Seven p16W melanoma-prone kindreds, on the other hand, had a total of 18 cases of melanoma, 4 cases of dysplastic naevi, and 9 cancers at other sites, but no cases of pancreatic cancer (26).

However, this excess of pancreatic cancer in FAMMM families may be attributed, in part, to ascertainment bias. Investigations of the tumor spectrum of the FAMMM syndrome revealed no excess of pancreatic tumors in a total of 15 FAMMM kindreds and 370 individuals diagnosed with melanoma (2830). The results from Australian kindreds (N.H., unpublished data) support this lack of excess of pancreatic cancer in FAMMM pedigrees: they found one case of pancreatic cancer in 11 families with p16M mutations (0.4 cases expected). This difference is not significant (95% CI, 0.49–24.56). Overall, there were slightly more cancers of all types among these families than among those with p16W CDKN2A mutations, but again, this difference was not significant.

It may be that the risk of pancreatic cancer is related to the position or type of CDKN2A mutation; in von Hippel Lindau disease kindreds, the risk of pheochromocytoma appears to be greatest in those with missense mutations in VHL (31), and in hereditary breast cancer, the risk of ovarian cancer may depend upon the position of the mutation along the BRCA1 gene (32).

CDKN2A Mutations in Melanoma Families Apparently Not Linked to 9p

Because of the high phenocopy rate, mutations have also been reported in families inconclusive for linkage to 9p (21,23). Some cases of melanoma observed in these families appear to be sporadic in origin and, in contrast to the definitively 9p-linked families, CDKN2A mutants in these unlinked kindreds did not always segregate with the melanomas.

Non-FAMMM Kindreds with CDKN2A Mutations

CDKN2A mutations are not restricted to the FAMMM syndrome. Interestingly, however, there are no reported mutations in families with multiple cancers that do not include melanoma. A family in which pancreatic cancer was the predominant cancer has been reported (33). A p16M missense mutation (Gly101Trp) was found in all affected family members, but only one individual was affected with melanoma (followed by pancreatic cancer). Sun et al. detected a Met53Ile CDKN2A mutation in a non-FAMMM family with an excessive number of cancers. One member of the family developed two cutaneous malignant melanomas during pregnancy, but no other individuals have had melanomas or dysplastic nevi. A p16M (deletion of amino acids 96–99) mutation was reported in a family with melanoma, nonsmall cell lung cancer, and squamous cell carcinoma of the head and neck (34). It appears that CDKN2A is highly penetrant for cutaneous malignant melanoma, and the absence of this cancer in a pedigree with multiple cases of other cancers is likely to imply that a CDKN2A mutation is not present in that family (W.F., unpublished data).

Somatic Mutations

CDKN2A is frequently homozygously deleted in cell lines derived from a number of tumor types (13,16,35). In melanoma cell lines where one copy of the gene is absent, the remaining copy is frequently mutated (7). The majority of point mutations observed are nonsense, missense, or frameshift mutations. Details of the coding effect of CDKN2A mutations can be found in a recent review by Pollock et al. (36). We have built upon the databases compiled by Pollock et al. (36) and Smith-Sorensen and Hovig (37) of CDKN2A somatic mutations in cell lines and in primary tumors, and present updated lists (to July 1996) in Tables 1 and 2, respectively.

Table 1 CDKN2A Somatic mutations identified to date (July 1996) in cell lines
Table 2 CDKN2A somatic mutations identified to date (July 1996) in primary tumors

It is interesting to note that alterations have been reported to occur in at least 70% of the possible 156 codons making up this small gene (deletions affecting more than one codon were counted only once). Moreover, only 11% of the germline mutations identified to date (Fig. 2) have been seen somatically (in either primary tumors or cell lines). There are several possible explanations for the latter phenomenon: (1) ascertainment bias—novel germline mutations may be detected in kindreds displaying different phenotypes than the families tested so far; (2) deleterious mutations—certain CDKN2A germline mutations may not be compatible with life; (3) somatic mutations reflect mutagenesis by a different carcinogen (i.e., ultraviolet radiation induces characteristic transitions and tandem base changes); and, (4) the lack of concordance between somatic and germline mutations may be attributed to chance.

Homozygous Deletions in Cell Lines

There have been numerous reports of homozygous deletion of the 9p21 region in cell lines derived from a wide variety of human tumors (7,35). CDKN2A has often (but not always) proven to be the target of these deletions. If a gene is the sole target of homozygous deletion, one would expect to find intragenic mutations in the DNA of cell lines without homozygous deletions. This criterion is not always met in the case of CDKN2A, however, which suggests the presence of another gene in the region that is responsible for the deletions observed; the neighboring CDKN2B gene is an obvious candidate. Microdeletions, aberrant CpG island methylation, or other regulatory mutations in CDKN2A not detected by the assays used by these investigators, may account for part of the reported absence of homozygous deletions (see above).

Cell lines with deletions of CDKN2A alone, CDKN2B alone, and with codeletion of the two genes have all been observed (2, 13,3845). CDKN2A is usually but not always included in the smallest region of homozygous deletion (2,38,39,46). This result can be interpreted in various ways: (1) the target gene is CDKN2A in some cases and CDKN2B in others; (2) both genes together are the target of the deletions; or (3) other tumor suppressor genes are present in the region. Jen et al. (38) favor the “double target” hypothesis because the protein products of the two genes appear to have analogous biochemical activities and the region of homozygous deletion most often contains both genes. A more definitive explanation awaits further functional analyses of the genes, especially CDKN2B.

Common Tumors with a Low Frequency of CDKN2A Mutations

colon carcinoma. CDKN2A mutations are rarely observed in colon cancer (7,13,16,38). Interestingly, aberrant 5′ CpG island methylation associated with loss of transcription of CDKN2A occurs frequently in both colon cancer cell lines and primary colon tumors (18). Of all the cell lines tested, those derived from colon primaries showed the highest frequency of de novo methylation (92%). CpG island methylation accompanied by transcriptional silencing of CDKN2A was also observed in the normal colonic mucosa of individuals with and without cancer (19). It is uncertain whether this uncommon situation represents a precancerous state or normal cell function. In general, these data point to hypermethylation as an alternative mechanism of CDKN2A inactivation in colon cancer.

breast cancer. CDKN2A is homozygously deleted in 60–65% of primary breast tumors and cell lines (7,16), however point mutations in this malignancy are rare (Tables 1 and 2). Of 24 primary breast carcinomas analyzed by Brenner et al., 58% showed LOH or allelic imbalance at 9p21–22, but only 1 of 21 had an intragenic mutation (47). Xu et al. screened 37 primary breast carcinomas and 5 cell lines for alterations in CDKN2A by single-strand conformation analysis (SSCA) (48). No mutations were found in any of the tumor samples but the gene was homozygously deleted in 2 of the 5 cell lines. Herman et al. reported CpG island methylation in 33% of breast cancer cell lines (18). Thus, it would appear that the primary mechanisms of CDKN2A inactivation in breast cancer are homozygous deletion and hypermethylation, but not point mutation.

ovarian carcinoma. LOH at 9p21 has been observed in 29–48% of ovarian tumors and cell lines (7,49); however, no tumor-specific CDKN2A mutations were detected in a total of 78 primary ovarian tumors analyzed by Campbell et al. (49) and Hatta et al. (50), and homozygous deletions were seen in only 2 tumors. It is not clear whether the high rate of 9p allelic loss in this malignancy is due to mechanisms of CDKN2A inactivation other than deletions and point mutations, or whether a different tumor suppressor gene is the target of LOH. There is an interesting parallel on chromosome 18q, where LOH is extremely frequent in colon cancer. At first, the gene DCC (51) was thought to be the sole target of the LOH, but it is now apparent that DPC4 and another related gene, JV-18-1, may also be mutated or otherwise inactivated in colorectal cancers (52,53). Similarly, perhaps other 9p21 cancer genes await identification.

The frequency of intragenic mutations in CDKN2A is relatively low in uncultured tumors. However, since there are other ways of abrogating p16CDKN2A function, the frequency of inactivation of CDKN2A in a given tumor type may in fact be higher than might be predicted from sequence analysis only. If this is the case, overall, CDKN2A may be somatically altered at a similar frequency to p53.

CDKN2A Mutations, Expression Levels, and Survival

Loss of p16CDKN2A expression may be related to invasiveness or metastatic potential rather than to tumor initiation. This is supported by the study of Reed et al. (54), who performed immunohistochemical analysis of p16CDKN2A expression on 103 melanocytic lesions ranging from atypical nevi to metastatic melanomas. p16CDKN2A expression was detected in 100% of atypical nevi and melanomas in situ and in 91% of primary invasive melanomas, but in only 56% of metastatic melanomas.

Nearly 80% of pancreatic cell lines or xenografts (55) and 37% of primary pancreatic adenocarcinomas (56) have CDKN2A mutations or deletions. The status of CDKN2A may be related to the prognosis of pancreatic cancer patients: mean survival was 13.5 months longer for individuals with CDKN2A mutation-negative tumors compared with those having mutation-positive tumors (p = 0.017) (57). It is interesting that LOH at 9p21 in breast cancers, on the other hand, may not confer an adverse prognosis (58). Thus it may be that the poorer survival in pancreatic carcinomas with CDKN2A mutations is either because CDKN2A mutations have a tumor-specific effect, or because in breast cancer, another gene is the target for the LOH and this gene does not have an adverse effect on survival.

Conclusions

Over the last 2 years, CDKN2A has been the subject of intensive research. It clearly plays a central role in the development of both hereditary and sporadic forms of melanoma. The gene is frequently altered in tumors of many different types, although its importance in most of these cancers can only be inferred. Future reviews of CDKN2A will hopefully focus on attempts to further dissect its function and to rectify the defects in vivo.

References

  1. Cairns P, Mao L, Merlo A, et al. (1994) Rates of p16 (MTS1) mutations in primary tumors with 9p loss. Science 265: 415–417.

    Article  PubMed  CAS  Google Scholar 

  2. Izumoto S, Arita N, Ohnishi T, Hiraga S, Taki T, Hayakawa T. (1995) Homozygous deletions of P16(INK4A)/MTS1 and P15 (INK4B)/MTS2 genes in glioma cells and primary glioma tissues. Cancer Lett. 97: 241–247.

    Article  PubMed  CAS  Google Scholar 

  3. Bergman W, Watson P, de Jong J, Lynch HT, Fusaro RM. (1990) Systemic cancer and the FAMMM syndrome. Br. J. Cancer 61: 932–936.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Hollstein M, Rice K, Greenblatt MS, et al. (1994) Database of p53 gene somatic mutations in human tumors and cell lines. Nucl. Acids Res. 22: 3551–3555.

    PubMed  CAS  Google Scholar 

  5. Li FP, Fraumeni JF, Jr, Mulvihill JJ, et al. (1988) A cancer family syndrome in twenty-four kindreds. Cancer Res. 48: 5358–5362.

    PubMed  CAS  Google Scholar 

  6. Fountain JW, Karayiorgou M, Ernstoff MS, et al. (1992) Homozygous deletions within human chromosome band 9p21 in melanoma. Proc. Natl. Acad. Sci. U.S.A. 89: 10557–10561.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Kamb A, Gruis NA, Weaver-Feldhaus J, et al. (1994) A cell cycle regulator potentially involved in genesis of many tumor types. Science 264: 436–440.

    Article  PubMed  CAS  Google Scholar 

  8. Cannon-Albright LA, Goldgar DE, Meyer LJ, et al. (1992) Assignment of a locus for familial melanoma, MLM, to chromosome 9p13–p22. Science 258: 1148–1152.

    Article  PubMed  CAS  Google Scholar 

  9. Goldstein AM, Dracopoli NC, Engelstein M, Fraser MC, Clark WH, Jr, Tucker MA. (1994) Linkage of cutaneous malignant melanoma/dysplastic nevi to chromosome 9p, and evidence for genetic heterogeneity. Am. J. Hum. Genet. 54: 489–496.

    PubMed  PubMed Central  CAS  Google Scholar 

  10. Nancarrow DJ, Mann GJ, Holland EA, et al. (1993) Confirmation of chromosome 9p linkage in familial melanoma. Am. J. Hum. Genet. 53: 936–942.

    PubMed  PubMed Central  CAS  Google Scholar 

  11. Gruis NA, Sandkuijl LA, Weber JL, et al. (1993) Linkage analysis in Dutch familial atypical multiple mole-melanoma (FAMMM) syndrome families. Effect of naevus count. Melanoma Res. 3: 271–277.

    PubMed  CAS  Google Scholar 

  12. MacGeoch C, Bishop JA, Bataille V, et al. (1994) Genetic heterogeneity in familial malignant melanoma. Hum. Mol. Genet. 3: 2195–2200.

    Article  PubMed  CAS  Google Scholar 

  13. Okamoto A, Demetrick DJ, Spillare EA, et al. (1994) Mutations and altered expression of p16INK4 in human cancer. Proc. Natl. Acad. Sci. U.S.A. 91: 11045–11049.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Ohta M, Nagai H, Shimizu M, et al. (1994) Rarity of somatic and germline mutations of the cyclin-dependent kinase 4 inhibitor gene, CDK4I, in melanoma. Cancer Res. 54: 5269–5272.

    PubMed  CAS  Google Scholar 

  15. Spruck CH, 3rd, Gonzalez-Zulueta M, Shibata A, et al. (1994) p16 gene in uncultured tumours. Nature 370: 183–184.

    Article  PubMed  Google Scholar 

  16. Cairns P, Polascik TJ, Eby Y, et al. (1995) Frequency of homozygous deletion at p16/CDKN2 in primary human tumours. Nature Genet. 11: 210–212.

    Article  PubMed  CAS  Google Scholar 

  17. Merlo A, Herman JG, Mao L, et al. (1995) 5′ CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers. Nature Med. 1: 686–692.

    Article  PubMed  CAS  Google Scholar 

  18. Herman JG, Merlo A, Mao L, et al. (1995) Inactivation of the CDKN2/p16/MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers. Cancer Res. 55: 4525–4530.

    PubMed  CAS  Google Scholar 

  19. Gonzalez-Zulueta M, Bender CM, Yang AS, et al. (1995) Methylation of the 5′ CpG island of the p16/CDKN2 tumor suppressor gene in normal and transformed human tissues correlates with gene silencing. Cancer Res. 55: 4531–4535.

    PubMed  CAS  Google Scholar 

  20. Reed AL, Califano J, Cairus P, Wesira WH, Jones RM, Koch W, Ahrendt S, Eby Y, Sewell D, Nawroe H, Bartek J, Sidransky D. (1996) High frequency of p16 (CDKN2/MTS-1/INK4A) inactivation in head and neck squamous cell carcinoma. Cancer Res. 56:3630–3633.

    PubMed  CAS  Google Scholar 

  21. Hussussian CJ, Struewing JP, Goldstein AM, et al. (1994) Germline p16 mutations in familial melanoma. Nature Genet. 8: 15–21.

    Article  PubMed  CAS  Google Scholar 

  22. Gruis NA, van der Velden PA, Sandkuijl LA, et al. (1995) Homozygotes for CDKN2 (p16) germline mutation in Dutch familial melanoma kindreds. Nature Genet. 10: 351–353.

    Article  PubMed  CAS  Google Scholar 

  23. Walker GJ, Hussussian CJ, Flore JF, et al. (1995) Mutations of the CDKN2/p16(INK4) gene in Australian melanoma kindreds. Hum Mol. Genet. 4: 1845–1852.

    Article  PubMed  CAS  Google Scholar 

  24. Sun S, Narod SA, Foulkes WD. (1996) A p16 mutation in a family with multiple cancers [abstract]. Eur J Hum Genet. 4: 13.

    Article  Google Scholar 

  25. Goldstein AM, Fraser MC, Struewing JP, et al. (1995) Increased risk of pancreatic cancer in melanoma-prone kindreds with p16INK4 mutations. N. Engl. J. Med. 333: 970–974.

    Article  PubMed  CAS  Google Scholar 

  26. Ciotti P, Strigini P, Bianchiscarra G. (1996) Familial melanoma and pancreatic cancer. N. Engl. J. Med. 334: 469–470.

    Article  PubMed  CAS  Google Scholar 

  27. Ranade K, Hussussian CJ, Sikorski RS, et al. (1995) Mutations associated with familial melanoma impair p16INK4 function. Nature Genet. 10: 114–116.

    Article  PubMed  CAS  Google Scholar 

  28. Lynch HT, Fusaro RM, Pester J, et al. (1981) Tumour spectrum in the FAMMM syndrome. Br. J. Cancer 44: 553–560.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Greene MH, Tucker MA, Clark WH, Jr, Kraemer KH, Elder DE, Fraser MC. (1987) Hereditary melanoma and the dysplastic nevus syndrome: The risk of cancers other than melanoma. J. Am. Acad. Dermatol. 16: 792–797.

    Article  PubMed  CAS  Google Scholar 

  30. Gutman M, Cnaan A, Inbar M, et al. (1991) Are malignant melanoma patients at higher risk for a second cancer? Cancer 68: 660–665.

    Article  PubMed  CAS  Google Scholar 

  31. Linehan WM, Lerman MI, Zbar B. (1995) Identification of the von Hippel-Lindau (VHL) gene. Its role in renal cancer. JAMA 273: 564–570.

    Article  PubMed  CAS  Google Scholar 

  32. Gayther SA, Warren W, Mazoyer S, et al. (1995) Germline mutations of the BRCA1 gene in breast and ovarian cancer families provide evidence for a genotype-phenotype correlation. Nature Genet. 11: 428–433.

    Article  PubMed  CAS  Google Scholar 

  33. Whelan AJ, Bartsch D, Goodfellow PJ. (1995) Brief report: a familial syndrome of pancreatic cancer and melanoma with a mutation in the CDKN2 tumor-suppressor gene. N. Engl. J. Med. 333: 975–977.

    Article  PubMed  CAS  Google Scholar 

  34. Yarbrough WG, Aprelikova O, Pei H, Olshan AF, Liu ET. (1996) Familial tumor syndrome associated with a germline non-functional p16(INK4a) allele. J. Natl. Cancer Inst. 88: 1489–1491.

    Article  PubMed  CAS  Google Scholar 

  35. Nobori T, Miura K, Wu DJ, Lois A, Takabayashi K, Carson DA. (1994) Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Nature 368: 753–756.

    Article  PubMed  CAS  Google Scholar 

  36. Pollock PM, Pearson JV, Hayward NK. (1996) Compilation of somatic mutations of the CDKN2 gene in human cancers—non-random distribution of base substitutions. Genes Chromosome Cancer 15: 77–88.

    Article  CAS  Google Scholar 

  37. Smith-Sorensen B, Hovig E. (1996) CDKN2A (P16(INK4A)) somatic and germline mutations. Hum. Mutat. 7: 294–303.

    Article  PubMed  CAS  Google Scholar 

  38. Jen J, Harper JW, Bigner SH, et al. (1994) Deletion of p16 and p15 genes in brain tumors. Cancer Res. 54: 6353–6358.

    PubMed  CAS  Google Scholar 

  39. Yoshida S, Todoroki T, Ichikawa Y, et al. (1995) Mutations of p16Ink4/CDKN2 and p15Ink4B/MTS2 genes in biliary tract cancers. Cancer Res. 55: 2756–2760.

    PubMed  CAS  Google Scholar 

  40. Luca M, Xie SH, Gutman M, Huang SY, Bareli M. (1995) Abnormalities in the CDKN2 (P16(INK4)/MTS-1) gene in human melanoma cells—relevance to tumor growth and metastasis. Oncogene 11: 1399–1402.

    PubMed  CAS  Google Scholar 

  41. Nakagawa K, Conrad NK, Williams JP, Johnson BE, Kelley MJ. (1995) Mechanism of inactivation of CDKN2 and MTS2 in non-small cell lung cancer and association with advanced stage. Oncogene 11: 1843–1851.

    PubMed  CAS  Google Scholar 

  42. Huang LY, Goodrow TL, Zhang SY, Kleinszanto AJP, Chang H, Ruggeri BA. (1996) Deletion and mutation analyses of the p16/MTS-1 tumor suppressor gene in human ductal pancreatic cancer reveals a higher frequency of abnormalities in tumor-derived cell lines than in primary ductal adenocarcinomas. Cancer Res. 56: 1137–1141.

    PubMed  CAS  Google Scholar 

  43. Zariwala M, Liu E, Xiong Y. (1996) Mutational analysis of the p16 family cyclin-dependent kinase inhibitors p15(INK4B) and p18(INK4C) in tumor-derived cell lines and primary tumors. Oncogene 12: 451–455.

    PubMed  CAS  Google Scholar 

  44. Naumann M, Savitskaia N, Eilert C, Schramm A, Kalthoff H, Schmiegel W. (1996) Frequent codeletion of p16/MTS1 and p15/MTS2 and genetic alterations in p16/MTS1 in pancreatic tumors. Gastroenterology 110: 1215–1224.

    Article  PubMed  CAS  Google Scholar 

  45. Miller CW, Aslo A, Campbell MJ, Kawamata N, Lampkin BC, Koeffler HP. (1996) Alterations of the p15, p16, and p18 genes in osteosarcoma. Cancer Genet. Cytogenet. 86: 136–142.

    Article  PubMed  CAS  Google Scholar 

  46. Wiest J, Frankin W, Otstot J, Anderson M. (1996) Identification of a novel region of homozygous deletion on chromosome 9p in squamous cell carcinoma of the lung [abstract]. Proc. Am. Assoc. Cancer Res. 37: 593.

    Google Scholar 

  47. Brenner AJ, Aldaz CM. (1995) Chromosome 9p allelic loss and p16/CDKN2 in breast cancer and evidence of p16 inactivation in immortal breast epithelial cells. Cancer Res. 55: 2892–2895.

    PubMed  CAS  Google Scholar 

  48. Xu L, Sgroi D, Sterner CJ, et al. (1994) Mutational analysis of CDKN2 (MTS1/p16ink4) in human breast carcinomas. Cancer Res. 54: 5262–5264.

    PubMed  CAS  Google Scholar 

  49. Campbell IG, Foulkes WD, Beynon G, Davis M, Englefield P. (1995) LOH and mutation analysis of CDKN2 in primary human ovarian cancers. Int. J. Cancer 63: 222–225.

    Article  PubMed  CAS  Google Scholar 

  50. Hatta Y, Hirama T, Takeuchi S, et al. (1995) Alterations of the p16 (MTS1) gene in testicular, ovarian, and endometrial malignancies. J Urol. 154: 1954–1957.

    Article  PubMed  CAS  Google Scholar 

  51. Fearon ER, Cho KR, Nigro JM, et al. (1990) Identification of a chromosome 18q gene that is altered in colorectal cancers. Science 247: 49–56.

    Article  PubMed  CAS  Google Scholar 

  52. Thiagalingam S, Lengauer C, Leach FS, et al. (1996) Evaluation of candidate tumour suppressor genes on chromosome 18 in colorectal cancers. Nature Genet. 13: 343–346.

    Article  PubMed  CAS  Google Scholar 

  53. Riggins GJ, Thiagalingam S, Rozenblum E, et al. (1996) Mad-related genes in the human. Nature Genet. 13: 347–349.

    Article  PubMed  CAS  Google Scholar 

  54. Reed JA, Loganzo F, Jr, Shea CR, et al. (1995) Loss of expression of the p16/cyclin-dependent kinase inhibitor 2 tumor suppressor gene in melanocytic lesions correlates with invasive stage of tumor progression. Cancer Res. 55: 2713–2718.

    PubMed  CAS  Google Scholar 

  55. Caldas C, Hahn SA, da Costa LT, et al. (1994) Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nature Genet. 8: 27–32.

    Article  PubMed  CAS  Google Scholar 

  56. Bartsch D, Shevlin DW, Tung WS, Kisker O, Wells SA, Goodfellow PJ. (1995) Frequent mutations of CDKN2 in primary pancreatic adenocarcinomas. Genes Chromosome Cancer 14: 189–195.

    Article  CAS  Google Scholar 

  57. Bartsch D, Shevlin DW, Callery MP, Norton JA, Wells SA, Goodfellow PJ. (1996) Reduced survival in patients with ductal pancreatic adenocarcinoma associated with CDKN2 mutation. J. Natl. Cancer Inst. 88: 680–682.

    Article  PubMed  CAS  Google Scholar 

  58. Eiriksdottir G, Sigurdsson A, Jonasson JG, et al. (1995) Loss of heterozygosity on chromosome 9 in human breast cancer—association with clinical variables and genetic changes at other chromosome regions. Int. J. Cancer 64: 378–382.

    Article  PubMed  CAS  Google Scholar 

  59. Holland EA, Beaton SC, Becker TM, et al. (1995) Analysis of the p16 gene, CDKN2, in 17 Australian melanoma kindreds. Oncogene 11: 2289–2294.

    PubMed  CAS  Google Scholar 

  60. Hansson J, Platz A, Linder S, Mansson BE, Lundqvist E, Inganas P, et al. (1996) Germine p16 mutations are rare in Swedish melanoma families [abstract]. Proc. Am. Assoc. Cancer Res. 37: 186.

    Google Scholar 

  61. Kamb A, Shattuck-Eidens D, Eeles R, et al. (1994) Analysis of the p16 gene (CDKN2) as a candidate for the chromosome 9p melanoma susceptibility locus. Nature Genet. 8: 23–26.

    Article  PubMed  CAS  Google Scholar 

  62. Liu L, Lassam NJ, Slingerland JM, et al. (1995) Germline p16INK4A mutation and protein dysfunction in a family with inherited melanoma. Oncogene 11: 405–412.

    PubMed  CAS  Google Scholar 

  63. Borg A, Johansson U, Johannsson O, et al. (1996) Novel germline p16 mutation in familial malignant melanoma in Southern Sweden. Cancer Res. 56: 2497–2500.

    PubMed  CAS  Google Scholar 

  64. Ueki K, Rubio MP, Ramesh V, et al. (1994) MTS1/CDKN2 gene mutations are rare in primary human astrocytomas with allelic loss of chromosome 9p. Hum. Mol. Genet. 3: 1841–1845.

    Article  PubMed  CAS  Google Scholar 

  65. Musgrove EA, Lilischkis R, Cornish AL, et al. (1995) Expression of the cyclin-dependent kinase inhibitors p16(INK4), p15(INK4B) and p21(WAF1/CIP1) in human breast cancer. Int. J. Cancer 63: 584–591.

    Article  PubMed  CAS  Google Scholar 

  66. Ohnishi H, Kawamura M, Ida K, et al. (1995) Homozygous deletions of p16/MTS1 gene are frequent but mutations are infrequent in childhood T-cell acute lymphoblastic leukemia. Blood 86: 1269–1275.

    PubMed  CAS  Google Scholar 

  67. Uzawa K, Suzuki H, Yokoe H, Tanzawa H, Sato K. (1995) Mutational state of p16/CDKN2 and VHL genes in squamous-cell carcinoma of the oral cavity. Int. J. Oncol. 7: 895–899.

    PubMed  CAS  Google Scholar 

  68. Calabro V, Strazzullo M, LaMantia G, Fedele M, Paulin C, Fusco A. (1996) Status and expression of the p16(INK4) gene in human thyroid-tumour cell lines. Int. J. Cancer 67: 29–34.

    Article  PubMed  CAS  Google Scholar 

  69. Nakamaki T, Kawamata N, Schwaller J, et al. (1995) Structural integrity of the cyclin-dependent kinase inhibitor genes, p15, p16 and p18 in myeloid leukaemias. Br. J. Haematol. 91: 139–149.

    Article  PubMed  CAS  Google Scholar 

  70. De Vos S, Miller CW, Takeuchi S, Gombart AF, Cho SK, Koeffler HP. (1995) Alterations of CDKN2 (p16) in non-smal cell lung cancer. Genes Chromosome Cancer 14: 164–170.

    Article  Google Scholar 

  71. Yang CH, Perentesis JP. (1996) Novel mutations of the CDKN2A gene in leukemia: Potential insight into the structural basis of CDKN2A action [abstract]. Proc Am Assoc Cancer Res 37: 590.

    Google Scholar 

  72. Okajima E, Fukuda T, Okita S, Tsutsumi M, Hirao Y, Konishi Y. (1996) Infrequent somatic alteration of p16/MTS1 in human primary superficial bladder cancers. Cancer Lett. 103: 227–231.

    Article  PubMed  CAS  Google Scholar 

  73. Platz A, Ringborg U, Lagerlof B, Lundqvist E, Sevigny P, Inganas M. (1996) Mutational analysis of the CDKN2 gene in metastases from patients with cutaneous malignant melanoma. Br. J. Cancer 73: 344–348.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Rasool O, Heyman M, Brandter LB, et al. (1995) p15ink4B and p16ink4 gene inactivation in acute lymphocytic leukemia. Blood 85: 3431–3436.

    PubMed  CAS  Google Scholar 

  75. Uchida T, Watanabe T, Kinoshita T, Murate T, Saito H, Hotta T. (1995) Mutational analysis of the CDKN2 (MTS1/p16ink4A) gene in primary B-cell lymphomas. Blood 86: 2724–2731.

    PubMed  CAS  Google Scholar 

  76. Esteve A, Martelplanche G, Sylla BS, Hollstein M, Hainaut P, Montesano R. (1996) Low frequency of p16/CDKN2 gene mutations in esophageal carcinomas. Int. J. Cancer 66: 301–304.

    Article  PubMed  CAS  Google Scholar 

  77. Ueki K, Ono Y, Hensen JW, Efird JT, von Deimling A, Louis DN. (1996) CDKN2/p16 or Rb alterations occur in the majority of glioblastomas and are inversely correlated. Cancer Res. 56: 150–153.

    PubMed  CAS  Google Scholar 

  78. Shimizu T, Sekiya T. (1995) Loss of heterozygosity at 9p21 loci and mutations of the MTS1 and MTS2 genes in human lung cancer. Int. J. Cancer 63: 616–620.

    Article  PubMed  CAS  Google Scholar 

  79. Konishi N, Hiasa Y, Tsuzuki T, et al. (1996) Detection of Rb, p16/CDKN2 and P15 (INK4B) gene alterations with immunohistochemical studies in human prostate carcinomas. Int. J. Oncol. 8: 107–112.

    PubMed  CAS  Google Scholar 

  80. Maelandsmo GM, Florenes VA, Hovig E, et al. (1996) Involvement of the pRb/p16/CDK4/cyclin D1 pathway in the tumorigenesis of sporadic malignant melanomas. Br. J. Cancer 73: 909–916.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Ohta M, Berd D, Shimizu M, et al. (1996) Deletion mapping of chromosome region 9p21–p22 surrounding the CDKN2 locus in melanoma. Int. J. Cancer 65: 762–767.

    Article  PubMed  CAS  Google Scholar 

  82. Rusin MR, Okamoto A, Chorazy M, et al. (1996) Intragenic mutations of the p16(INK4), p15(INK4B) and p18 genes in primary non-small-cell lung cancers. Int. J. Cancer 65: 734–739.

    Article  PubMed  CAS  Google Scholar 

  83. Haider MA, Cao XB, Manshouri T, Chan LL, Glassman A, Kantarjian HM. (1995) p16(INK4A) and p15(INK4B) gene deletions in primary leukemias. Blood 86: 311–315.

    Google Scholar 

  84. Kyritsis AP, Zhang BH, Zhang W, et al. (1996) Mutations of the p16 gene in gliomas. Oncogene 12: 63–67.

    PubMed  CAS  Google Scholar 

  85. Berns EMJJ, Klijn JGM, Smid M, Vanstaveren IL, Gruis NA, Foekens JA. (1995) Infrequent CDKN2 (MTS1/p16) gene alterations in human primary breast cancer. Br. J. Cancer 72: 964–967.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Nakao M, Yokota S, Kaneko H, et al. (1996) Alterations of CDKN2 gene structure in childhood acute lymphoblastic leukemia—mutations of CDKN2 are observed preferentially in T lineage. Leukemia 10: 249–254.

    PubMed  CAS  Google Scholar 

  87. Tung WS, Shevlin DW, Bartsch D, Norton JA, Wells SA, Goodfellow PJ. (1996) Infrequent CDKN2 mutation in human differentiated thyroid cancers. Mol. Carcinog. 15: 5–10.

    Article  PubMed  CAS  Google Scholar 

  88. Qin LX, Tang ZY, Liu KD, et al. (1996) Alterations of CDKN2(p16/MTS1) exon 2 in human hepatocellular carcinoma. Oncol. Rep. 3: 405–408.

    PubMed  CAS  Google Scholar 

  89. Miyamoto H, Kubota Y, Fujinami K, et al. (1995) Infrequent somatic mutations of the p16 and p15 genes in human bladder cancer—p16 mutations occur only in low-grade and superficial bladder cancers. Oncol. Res. 7: 327–330.

    PubMed  CAS  Google Scholar 

  90. Cayuela JM, Madani A, Sanhes L, Stern MH, Sigaux F. (1996) Multiple tumor-suppressor gene 1 inactivation is the most frequent genetic alteration in T-cell acute lymphoblastic leukemia. Blood 87: 2180–2186.

    PubMed  CAS  Google Scholar 

  91. Gonzalez-Zulueta M, Shibata A, Ohneseit PF, et al. (1995) High frequency of chromosome 9p allelic loss and CDKN2 tumor suppressor gene alterations in squamous cell carcinoma of the bladder. J. Natl. Cancer. Inst. 87: 1383–1393.

    Article  PubMed  CAS  Google Scholar 

  92. Sonoda Y, Yoshimoto T, Sekiya T. (1995) Homozygous deletion of the MTS1/p16 and MTS2/p15 genes and amplification of the CDK4 gene in glioma. Oncogene 11: 2145–2149.

    PubMed  CAS  Google Scholar 

  93. Igaki H, Sasaki H, Tachimori Y, et al. (1995) Mutation frequency of the p16/CDKN2 gene in primary cancers in the upper digestive tract. Cancer Res. 55: 3421–3423.

    PubMed  CAS  Google Scholar 

Download references

Acknowledgments

We thank Sophie Sun for her contribution to the tables and figures. WDF is supported by grants from Fonds de la recherche en Santé du Québec and the Cancer Research Society of Canada.

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Foulkes, W.D., Flanders, T.Y., Pollock, P.M. et al. The CDKN2A (p16) Gene and Human Cancer. Mol Med 3, 5–20 (1997). https://doi.org/10.1007/BF03401664

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/BF03401664

Keywords