Skip to main content
  • Commentary
  • Published:

E-cadherin and loss of heterozygosity at chromosome 16 in breast carcinogenesis: different genetic pathways in ductal and lobular breast cancer?

Abstract

Loss of heterozygosity at the long arm of chromosome 16 is one of the most frequent genetic events in breast cancer. In the search for tumour suppressor genes that are the target of loss of heterozygosity at 16q, the E-cadherin gene CDH1 was unveiled by the identification of truncating mutations in the retained copy. However, only lobular tumours showed E-cadherin mutations. Whereas investigations are still devoted to finding the target genes in the more frequent ductal breast cancers, other studies suspect the E-cadherin gene to also be the target in this tumour type. The present article discusses the plausibility of those two lines of thought.

Introduction

The homotypic cellular adhesion molecule E-cadherin is one of the most vital components in the cell. It is implicated as a key player in different cellular processes including development, morphology, polarity, migration and tissue integrity [1]. E-cadherin is a glycoprotein with an extracellular domain that interacts with E-cadherin molecules on adjacent cells, thereby establishing adhesion between epithelial cells. The intracellular domain is associated with a complex of proteins called catenins, which anchor E-cadherin to the actin cytoskeleton.

In various carcinomas, plasma membrane associated E-cadherin protein expression is decreased or even absent. There is transcriptional regulation of this molecule for which a number of factors have been implicated, including promotor hypermethylation [2,3], and several proteins that regulate E-cadherin transcription, especially Snail-1 [4], SIP-1 [5] and integrin-linked kinase [6]. Mutational inactivation of the E-cadherin gene CDH1 has been reported in diffuse gastric cancer and lobular breast cancer [7]. Both these tumour types have a characteristic diffuse growth pattern with loss of cellular coherence that is in accordance with the adhesion function of the absent E-cadherin protein.

The wild type CDH1 allele is missing in most lobular tumours due to loss of heterozygosity (LOH) at chromosome 16q [8], thereby presenting a classical example of Knudson's two-hit hypothesis on the inactivation of tumour suppressor genes. The more frequent ductal breast carcinomas also show frequent LOH at 16q; however, these tumours do not have mutational inactivation of the retained CDH1 allele [9]. Given the importance and widespread involvement of E-cadherin in tumorigenic processes, it is tempting to assume that a decrease in E-cadherin activity in ductal carcinomas is selected for and is reflected by LOH at 16q. Indeed, haploinsufficiency has now been acknowledged as a true mechanism for tumour suppressor gene inactivation [10]. However, other genes at 16q could be targets of LOH in ductal tumours, justifying ongoing gene hunts.

LOH at 16q in ductal and lobular breast cancer

LOH at 16q is the second most frequent somatic genetic event in breast cancer. This event occurs in about 50% of all ductal carcinomas [11] and is slightly more frequent in lobular breast cancer [8]. To confirm E-cadherin as the target of LOH in ductal carcinoma, it is important to distinguish physical loss and mitotic recombination [12,13]. Only the first LOH event could theoretically lead to haplo-insufficiency of CDH1. This seems unlikely, however, since there are so many complex mechanisms for regulation of CDH1 transcription that are often part of feedback loops [14]. Furthermore, our observations (unpublished data) on LOH at 16q in breast cancer indicate that both mechanisms for LOH are operative.

E-cadherin protein expression in ductal and lobular breast cancer

The complete absence of E-cadherin plasma membrane associated protein expression as detected by immunohistochemistry is so unambiguous that pathologists use this immunostaining to confirm their diagnosis of lobular breast cancer. We have stained a series of 86 breast carcinomas with known E-cadherin mutation status for E-cadherin protein expression [15]. Complete loss of protein was found in all lobular tumours with mutational inactivation of E-cadherin (n = 21). In addition, we were unable to detect E-cadherin in 11 lobular tumours in which no mutation had been identified. This is probably due to insensitivity of the mutation detection method or because other mechanisms of inactivation (e.g. methylation) were active. Remarkably, six cases of lobular breast cancer without a detectable E-cadherin mutation were positive for E-cadherin immunostaining. Of the 48 ductal breast cancers tested, 37% showed a decrease in but never a complete absence of E-cadherin protein expression. If E-cadherin was the target of LOH at 16q in ductal breast cancer, one would expect a strong association between LOH at 16q and decreased E-cadherin expression. However, the percentage of LOH at 16q in tumours with and without E-cadherin decrease was equal. It therefore seems unlikely that LOH at 16q is associated with a decrease in E-cadherin expression in ductal breast cancer. Also, in ductal carcinoma in situ (DCIS), there is no association between LOH at 16q and a decrease in E-cadherin expression. This can be derived from our combined data on LOH and E-cadherin immunohistochemistry, which was available for 62 cases of pure DCIS [16,17].

The adhesion function of E-cadherin is strongly indicative of a function in invasion suppression and this has indeed been shown in vitro [18] and in mouse models [19]. Investigation in primary tumours showed that this function translates well into metastatic potential [20]. There is no significant correlation between LOH at 16q in breast cancer and metastatic potential [11], however, which further contradicts an association between E-cadherin and LOH at 16q.

A breast cancer progression model

The invasion suppressor function of E-cadherin is very obvious, but not in concordance with our earlier finding [16] that mutational inactivation of both CDH1 alleles through LOH and truncating mutations occurs in the prein-vasive stage in lobular carcinoma in situ (LCIS), a tumour stage that involves proliferation but not dissemination. We showed that the same mutation and LOH at 16q was present in the invasive tumour and the adjacent LCIS [16]. E-cadherin may thus play a role in invasive capacity, but more data suggest other signalling mechanisms may be involved in earlier tumorigenic processes, especially cellular proliferation. Indeed, E-cadherin is implicated in several signalling pathways: Wnt, Rho/Rac and p27Kip1, which are involved in transcriptional activation, actin cytoskeleton reorganisation and contact inhibition, respectively [14].

LOH at 16q also occurs in the preinvasive stage, predominantly in grade I DCIS and in LCIS [17,21]. These observations and data in the literature led to a progression model of breast cancer (Fig. 1) based on early genetic alterations [17]. A similar multistep model for breast carcinogenesis has been proposed by Lakhani [22]. The model by Vos et al. [17] is less well defined than the Vogelstein model for colorectal cancer, because the latter is based on specific tumour stages and gene mutations. Similar stages are not defined for breast cancer and most of the genes involved are not yet identified. However, somatic genetic alterations in DCIS, LCIS and invasive carcinoma indicate that breast cancer progression is also based on the accumulation of genetic alterations. LOH at 16q in grade I DCIS and LCIS led us to suggest that grade I DCIS may be a precursor for LCIS. Loss of E-cad-herin further determines the histological fate of the tumour. Loss of chromosome 16q in grade I ductal and lobular breast cancer also lead Roylance et al. to speculate that these apparently morphological different tumours have a common molecular origin [23]. The observation of mixed populations of tumour cells, LCIS adjacent to DCIS, infiltrating lobular carcinoma and infiltrating ductal carcinoma (Fig. 2) supports this model, as well as investigations by Buerger et al. [24] on comparative genomic hybridisation of different tumour populations within the same lesion.

Figure 1
figure 1

Model of breast cancer progression. Grade II tumours are omitted for simplification. Two possible pathways may result in lobular carcinoma: 1, direct; or 2, via well-differentiated ductal carcinoma in situ (CIS). IDC, infiltrating ductal carcinoma; ILC, infiltrating lobular carcinoma; LOH, loss of heterozygosity.

Figure 2
figure 2

E-cadherin staining of mixed populations of (a) ductal carcinoma in situ (DCIS) and lobular carcinoma in situ (LCIS), and (b) infiltrating ductal carcinoma (IDC) and infiltrating lobular carcinoma (ILC). The E-cadherin staining was performed with antibody HECD-1 and distinguishes ductal from lobular tumour cells. Courtesy of Dr CBJ Vos.

E-cadherin germline mutations and lack of LOH in gastric cancer

The identification of somatic E-cadherin mutations in breast and gastric cancer received less attention than the report on two Maori families with diffuse gastric cancer attributed to germline transmission of truncating mutations in CDH1 [25]. Although lobular breast cancer was expected, none was registered in these Maori families or in others reported on later [26]. Examination of CDH1 in 65 patients with LCIS revealed no germline mutations [27]. Loss of one CDH1 allele apparently gives an increased risk only for gastric cancer, both hereditary and sporadic.

Remarkably, at the level of LOH, there is also a substantial difference between breast and gastric tumours. In diffuse gastric tumours, the wild type CDH1 allele is inactivated not by LOH at 16q, but by promotor methylation [28]. This marked difference in general genetic mechanism may reflect a difference in the role of the tumour suppressor gene. Whereas the loss of E-cadherin is a rate-limiting factor in gastric cancer, in breast cancer it probably plays a role in a later stage and it determines the histological subtype.

Conclusions

LOH at chromosome arm 16q in breast cancer is a frequent event, occurring in at least 50% of breast cancer cases. In lobular breast cancer, a histological minority comprising 5–10% of all breast cancers, the E-cadherin gene is the target of this somatic genetic event. In ductal breast cancer it is unlikely that CDH1 is the target tumour suppressor gene, and other genes therefore remain to be identified. Classical LOH mapping efforts have not been successful in the identification of these target genes at chromosome 16, or other genes in other tumour types, and we therefore need to apply different high-throughput screening methods to identify these remaining genes.

The E-cadherin gene has many different functions, even in carcinogenesis, given its involvement in early lesions and metastasis, hereditary and sporadic tumours, and numerous different tumour types. To elucidate whether this remarkable diversity indicates true separate activities or is a reflection of this protein's central role in cellular processes will be a challenging task for cellular biologists, geneticists and oncology researchers together.

Abbreviations

CDH1:

E-cadherin gene

DCIS:

ductal carcinoma in situ

LCIS:

lobular carcinoma in situ

LOH:

loss of heterozygosity.

References

  1. Vleminckx K, Kemler R: Cadherins and tissue formation: integrating adhesion and signaling. Bioessays. 1999, 21: 211-220. 10.1002/(SICI)1521-1878(199903)21:3<211::AID-BIES5>3.0.CO;2-P.

    Article  CAS  PubMed  Google Scholar 

  2. Yoshiura K, Kanai Y, Ochiai A, Shimoyama Y, Sugimura T, Hiro-hashi S: Silencing of the E-cadherin invasion-suppressor gene by CpG methylation in human carcinomas. Proc Natl Acad Sci USA. 1995, 92: 7416-7419.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Graff JR, Herman JG, Lapidus RG, Chopra H, Xu R, Jarrard DF, Isaacs WB, Pitha PM, Davidson NE, Baylin SB: E-cadherin expression is silenced by DNA hypermethylation in human breast and prostate carcinomas. Cancer Res. 1995, 55: 5195-5199.

    CAS  PubMed  Google Scholar 

  4. Batlle E, Sancho E, Franci C, Dominguez D, Monfar M, Baulida J, Garcia DH: The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol. 2000, 2: 84-89. 10.1038/35000034.

    Article  CAS  PubMed  Google Scholar 

  5. Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L, Bruyneel E, Mareel M, Huylebroeck D, Van Roy F: The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell. 2001, 7: 1267-1278. 10.1016/S1097-2765(01)00260-X.

    Article  CAS  PubMed  Google Scholar 

  6. Tan C, Costello P, Sanghera J, Dominguez D, Baulida J, De Herreros AG, Dedhar S: Inhibition of integrin linked kinase (ILK) suppresses beta-catenin–Lef/Tcf-dependent transcription and expression of the E-cadherin repressor, snail, in APC-/- human colon carcinoma cells. Oncogene. 2001, 20: 133-140. 10.1038/sj.onc.1204052.

    Article  CAS  PubMed  Google Scholar 

  7. Berx G, Becker KF, Hofler H, Van Roy F: Mutations of the human E-cadherin (CDH1) gene. Hum Mutat. 1998, 12: 226-237. 10.1002/(SICI)1098-1004(1998)12:4<226::AID-HUMU2>3.0.CO;2-D.

    Article  CAS  PubMed  Google Scholar 

  8. Berx G, Cleton-Jansen AM, Strumane K, De Leeuw WJF, Nollet F, Van Roy F, Cornelisse C: E-cadherin is inactivated in a majority of invasive human lobular breast cancers by truncation mutations throughout its extracellular domain. Oncogene. 1996, 13: 1919-1925.

    CAS  PubMed  Google Scholar 

  9. Berx G, Cleton-Jansen AM, Nollet F, De Leeuw WJF, Van de Vijver MJ, Cornelisse C, Van Roy F: E-cadherin is a tumour invasion suppressor gene mutated in human lobular breast cancers. EMBO J. 1995, 14: 6107-6115.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Jones PA, Laird PW: Cancer epigenetics comes of age. Nat Genet. 1999, 21: 163-167. 10.1038/5947.

    Article  CAS  PubMed  Google Scholar 

  11. Cleton-Jansen AM, Callen DF, Seshadri R, Goldup S, McCallum B, Crawford J, Powell JA, Settasatian C, van Beerendonk H, Moer-land EW, Smit VT, Harris WH, Millis R, Morgan NV, Barnes D, Mathew CG, Cornelisse CJ: Loss of heterozygosity mapping at chromosome arm 16q in 712 breast tumors reveals factors that influence delineation of candidate regions. Cancer Res. 2001, 61: 1171-1177.

    CAS  PubMed  Google Scholar 

  12. Tischfield JA: Loss of heterozygosity or: how I learned to stop worrying and love mitotic recombination. Am J Hum Genet. 1997, 61: 995-999. 10.1086/301617.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Devilee P, Cleton-Jansen AM, Cornelisse CJ: Ever since Knudson. Trends Genet. 2001, 17: 569-573. 10.1016/S0168-9525(01)02416-7.

    Article  CAS  PubMed  Google Scholar 

  14. Beavon IR: Regulation of E-cadherin: does hypoxia initiate the metastatic cascade?. Mol Pathol. 1999, 52: 179-188.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. De Leeuw WJ, Berx G, Vos CB, Peterse JL, Van de Vijver MJ, Litvinov S, Van Roy F, Cornelisse CJ, Cleton-Jansen AM: Simultaneous loss of E-cadherin and catenins in invasive lobular breast cancer and lobular carcinoma in situ. J Pathol. 1997, 183: 404-411. 10.1002/(SICI)1096-9896(199712)183:4<404::AID-PATH1148>3.0.CO;2-9.

    Article  CAS  PubMed  Google Scholar 

  16. Vos CB, Cleton-Jansen AM, Berx G, De Leeuw WJ, ter Haar NT, Van Roy F, Cornelisse CJ, Peterse JL, Van de Vijver MJ: E-cadherin inactivation in lobular carcinoma in situ of the breast: an early event in tumorigenesis. Br J Cancer. 1997, 76: 1131-1133.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Vos CBJ, ter Haar NT, Rosenberg C, Peterse JL, Cleton-Jansen A-M, Cornelisse CJ, Van de Vijver MJ: Genetic alterations on chromosome 16 and 17 are important features of ductal carcinoma in situ of the breast and are associated with histological type. Br J Cancer. 1999, 81: 1410-1418. 10.1038/sj.bjc.6693372.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Vleminckx K, Vakaet L, Mareel M, Fiers W, Van Roy F: Genetic manipulations of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell. 1991, 66: 107-119.

    Article  CAS  PubMed  Google Scholar 

  19. Perl AK, Wilgenbus P, Dahl U, Semb H, Christofori G: A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature. 1998, 392: 190-193. 10.1038/32433.

    Article  CAS  PubMed  Google Scholar 

  20. Birchmeier W, Behrens J: Cadherin expression in carcinomas: Role in the formation of cell junctions and the prevention of invasiveness. Biochim Biophys Acta. 1994, 1198: 11-26. 10.1016/0304-419X(94)90003-5.

    CAS  PubMed  Google Scholar 

  21. Lakhani SR, Collins N, Sloane JP, Stratton MR: Loss of heterozy-gosity in lobular carcinoma in situ of the breast. J Clin Pathol. 1995, 48M: M74-M78.

    Google Scholar 

  22. Lakhani SR: The transition from hyperplasia to invasive carcinoma of the breast. J Pathol. 1999, 187: 272-278. 10.1002/(SICI)1096-9896(199902)187:3<272::AID-PATH265>3.0.CO;2-2.

    Article  CAS  PubMed  Google Scholar 

  23. Roylance R, Gorman P, Harris W, Liebmann R, Barnes D, Hanby A, Sheer D: Comparative genomic hybridization of breast tumors stratified by histological grade reveals new insights into the biological progression of breast cancer. Cancer Res. 1999, 59: 1433-1436.

    CAS  PubMed  Google Scholar 

  24. Buerger H, Simon R, Schafer KL, Diallo R, Littmann R, Poremba C, Van Diest PJ, Dockhorn-Dworniczak B, Bocker W: Genetic relation of lobular carcinoma in situ, ductal carcinoma in situ, and associated invasive carcinoma of the breast. Mol Pathol. 2000, 53: 118-121. 10.1136/mp.53.3.118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Guilford P, Hopkins J, Harraway J, McLeod M, McLeod N, Harawira P, Taite H, Scoular R, Miller A, Reeve AE: E-cadherin germline mutations in familial gastric cancer. Nature. 1998, 392: 402-405. 10.1038/32918.

    Article  CAS  PubMed  Google Scholar 

  26. Richards FM, McKee SA, Rajpar MH, Cole TR, Evans DG, Jankowski JA, McKeown C, Sanders DS, Maher ER: Germline E-cadherin gene (CDH1) mutations predispose to familial gastric cancer and colorectal cancer. Hum Mol Genet. 1999, 8: 607-610. 10.1093/hmg/8.4.607.

    Article  CAS  PubMed  Google Scholar 

  27. Rahman N, Stone JG, Coleman G, Gusterson B, Seal S, Marossy A, Lakhani SR, Ward A, Nash A, McKinna A, A'Hern R, Stratton MR, Houlston RS: Lobular carcinoma in situ of the breast is not caused by constitutional mutations in the E-cadherin gene. Br J Cancer. 2000, 82: 568-570. 10.1054/bjoc.1999.0965.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Grady WM, Willis J, Guilford PJ, Dunbier AK, Toro TT, Lynch H, Wiesner G, Ferguson K, Eng C, Park JG, Kim SJ, Markowitz S: Methylation of the CDH1 promoter as the second genetic hit in hereditary diffuse gastric cancer. Nat Genet. 2000, 26: 16-17. 10.1038/79120.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The author wishes to acknowledge Dr CJ Cornelisse, Dr PCW Hogendoorn, and Dr MJ van de Vijver for helpful discussions, Dr C Vos for providing Figure 2, and Dr C Vos and Dr E Robanus Maandag for providing the data on DCIS immunohistochemistry.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anne-Marie Cleton-Jansen.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Cleton-Jansen, AM. E-cadherin and loss of heterozygosity at chromosome 16 in breast carcinogenesis: different genetic pathways in ductal and lobular breast cancer?. Breast Cancer Res 4, 5 (2001). https://doi.org/10.1186/bcr416

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/bcr416

Keywords