Elsevier

Oral Oncology

Volume 72, September 2017, Pages 7-16
Oral Oncology

Review
Relevance of chromosomal band 11q13 in oral carcinogenesis: An update of current knowledge

https://doi.org/10.1016/j.oraloncology.2017.04.016Get rights and content

Highlights

  • 11q13 amplification is a key event in oral carcinogenesis.

  • We examine the 11q13 amplicon and its amplicon cores and drivers in OSCC.

  • We review the oncogenic implications of oncogenes in the 11q13 amplicon.

  • We analyze the therapeutic targeting of chromosome band 11q13 in HNSCC and OSCC.

  • We discuss future research lines.

Abstract

An important event in oral carcinogenesis is the amplification of chromosomal band 11q13, in which numerous oncogenes and some tumor-suppressor genes are localized and frequently co-amplified during the malignant transformation of oral epithelium. The objectives of this study were to review published data on the involvement of 11q13 amplification in oral cancer, to provide an update on novel concepts and terminology related to gene amplification, and to explore the composition of the 11q13 amplicon in OSCC, including its most relevant amplicon cores and potential drivers. We report on the critical oncogenes and tumor-suppressor genes in 11q13 that may play a major role in oral cancer, focusing on their functions, on the characteristics acquired by their amplification, and on their clinicopathological implications. Finally, we discuss the possible usefulness of the 11q13 region as a therapeutic target in oral cancer.

Introduction

DNA copy number aberrations [1] in the form of microscopic (>500 kbp) or submicroscopic (<500 kbp) genetic material gains or losses represent a major genetic disorder that can promote cancer development [2]. The aberrations are frequently observed as the net gain or loss of a complete chromosome (aneuploidy) or as the partial gain or loss of a chromosome, producing an amplification or deletion, respectively, of genetic material [1], [3], [4]. Gene amplification is defined as an increase in copy number in a restricted region of a chromosome arm [4] and is a common mechanism for oncogenesis, which is frequently associated with the overexpression of amplified genes [4]. Gene amplification also has programmed and strictly regulated physiological functions during development, as in Drosophila oogenesis, the development of salivary glands of Sciara Cophrophila larvae, or chicken myogenesis [5], [6]. It is considered to begin with a double-strand DNA break [4], while the aberrant progress of the cell through its cycle would likely require the evasion of some robust checkpoints, including the p53-mediated checkpoint [4], [7], [8]. The most plausible hypotheses to explain the initial double-strand DNA break have related it to fragile chromosome sites, telemetric dysfunction, or DNA replication errors [4]. Once established, amplification can appear in three different forms: extrachromosomal copies, also known as “double minutes”; intrachromosomal copies, forming homogeneously stained regions (HSRs); and copies dispersed throughout the genome [4], [6], [9]. The different forms of presentation can coexist in the same cell, which often carries >5 copies, with some reports of >500 copies [6].

The amplification of a region frequently produces the expression of genes associated with human tumor development. The degree of causal relationship between amplification and cancer development depends on the amplified region and tumor type. Thus, the alteration proto-oncogene v-myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog (MYCN), located in 2p24, amplified in neuroblastomata, is known to cause neuroblastoma development [9]. In contrast, the amplification of chromosome band 11q13 frequently houses various co-amplified genes and is considered a prime example of the complexity of amplification in cancer [4], [9], [10]. The identification and quantification of amplified DNA in tumor cells permits the isolation of new candidate oncogenes involved in cell growth regulation and possibly in tumor development. Comparative genome hybridization (CGH), fluorescent “in situ” hybridization (FISH), Quantitative microsatellite analysis (QuMA), BAC end sequencing, and digital karyotyping are the most widely applied techniques for mapping and measuring amplification [4]. Increases in the resolution of amplification mapping techniques have led to the development of specific concepts related to amplification, including: amplicon, i.e., a focal DNA region with copy number increase; and amplicon driver, i.e., an amplified gene within an amplicon, whose expression confers advantages to the host cell and contributes to the maintenance of the malignant phenotype [11]. Recent studies demonstrated that an amplicon can house more than one amplicon driver [11], [12]. However, the characterization of a gene as an amplicon driver is challenging, because the transcriptional activation of some oncogenes is not regulated by the amplification mechanism alone. For instance, CCND1 amplification in 11q13 is an essential mechanism for regulating overexpression of cyclin D1 protein in oral squamous cell carcinoma (OSCC); however, this overexpression is also regulated by other mechanisms such as mitogen-activated protein kinase (MAPK) or phosphatidylinositol 3-kinase (PI3K) signaling pathways, which are often altered in oral cancer, hampering identification of CCND1 as driver in the 11q13 amplicon. Another novel amplification-related concept is the amplicon core, i.e., the minimum unit within an amplicon that can be amplified independently from other regions in the same amplicon [11].

Genetic material amplification has diagnostic and prognostic implications and can serve as a therapeutic target, while research on MYC, erb-b2 receptor tyrosine kinase 2 (ERBB2), epidermal growth factor receptor (EGFR), and CCND1 genes appears especially promising. Various publications have described gene amplification as an acquired drug-resistance mechanism. Technological advances over the past decade have achieved the genomic characterization of 11q13 region amplification, which is prevalent in numerous tumor types, including cancers of breast (15–20%), ovary (16%), bladder (11%), lung (9%), and pancreas (14–25%), melanoma (9–44%), and esophagus squamous cell (33%) and head-and-neck (30–62%) carcinomas [11]. It is of particular importance in OSCC, in which amplification of chromosome band 11q13 is highly frequent and has major clinicopathological repercussions.

The objectives of this study were: to review published data on chromosome band 11q13 amplification in human cancer, particularly in OSCC; to provide an update on key terminology and concepts; and to explore the composition of 11q13 amplicon in OSCC, including its most relevant cores and potential drivers. We report on the critical oncogenes and tumor-suppressor genes located in 11q13, which may play a major role in oral cancer, describing their functions, the characteristics conferred by their amplification, and the clinicopathological implications. Finally, we evaluate the possible usefulness of the 11q13 region as a therapeutic target in oral cancer.

Locus 11q13 is one of the most frequently amplified and important chromosomal regions in human cancer. The 11q13 amplicon is formed by long sequences of amplified DNA. Almost all of its genes can be simultaneously co-amplified, and it often contains neighboring co-amplified genes, making elucidation of its role in cancer highly complex [4], [9], [10]. The amplification values of 11q13 are frequently low (3–10 copies) [9], with the amplified DNA localized intrachromosomally as an HSR, and all copies are usually, although not always, maintained in the original chromosomal region of 11q13 [4], [6], [13].

11q13 amplification was first reported around three decades ago in fibroblast growth factor 3 (FGF3) gene (also known as INT2) in breast cancer [14] and was subsequently described in other cancers, including squamous cell carcinomas [15]. Shortly afterwards, fibroblast growth factor 4 (FGF4) gene (HSR1), also mapped in band 11q13, was found to be co-amplified with FGF3 in most tumors with 11q13 amplification, including squamous cell carcinomas [16], [17]. However, despite the co-amplification of FGF3 and FGF4, the protein expression of these genes is frequently low or undetectable [10]. This has prompted the search for the true driver genes of 11q13, i.e., those that possess the capacity to overexpress proteins and offer the host cells an oncogenic advantage. Thus, CCND1 gene [18], which is invariably found to be amplified and overexpressed in squamous cell cancer [19], has been proposed as 11q13 amplicon driver [11]. It encodes cyclin D1 protein, which promotes G1/S transition in the cell cycle, it is overexpressed in numerous tumors [20] and it plays a key role in OSCC development [21]. Likewise, CTTN gene (aka EMS1), which can be co-amplified alongside its neighbor CCND1 in 11q13 and overexpressed in various human cancers [22], also appears to play a major role in tumor development. Other genes mapped in 11q13-q14 (EMSY, p21 (RAC1) activated kinase 1 [PAK1], and GRB2 associated binding protein 2 [GAB2]) may also confer selective advantages to cells in which they are amplified [11]. It appears likely that other co-amplified genes are involved in tumor development, underscoring the complexity of 11q13 amplicon. In the late 1990s, comparative genomic hybridization (CGH) studies observed the independent amplification of genes in different loci mapped in 11q13, not only neighboring genes, and demonstrated the presence of four distinct cores within the amplicon [11]. A more precise mapping of these cores in tumor tissue has recently been achieved using high-resolution microarray-based CGH, revealing the composition and borders of the 11q13 amplicon [11]. The author reported that Core 1 (66.4–67.3 Mb) contained genes with a possible role in human cancer, e.g., RNA binding motif protein 4 (RBM4), aryl hydrocarbon receptor interacting protein (AIP), or cyclin dependent kinase 2 associated protein 2 (CDK2AP2), as did Core 2 (68.8–70.3 Mb), e.g., CCND1, CTTN, Fas associated via death domain (FADD), oral cancer overexpressed 1 (ORAOV1) or FGF3/4, some of which (CCND1, CTTN, and FADD) have been proposed as potential amplicon drivers. Core 3 (75.4–76.4 Mb) was also found to contain relevant genes, e.g., Wnt family member 11 (WNT11) and EMSY, and the latter is also a candidate amplicon driver. Interestingly, Core 3 houses UV radiation resistance associated (UVRAG) gene, which has been attributed with tumor-suppressor activity, because it is an essential component of the Beclin1-PI3KC3 complex, an important tumor cell growth and autophagy signaling checkpoint [23]. In other words, the amplification patterns of genes in the 11q13 amplicon do not necessarily have oncogenic functions. Finally, core 4 (77.0–79.2), which extends to region 11q14.1, includes PAK1 and GAB2 genes, both proposed as possible amplicon drivers. Characteristic 11q13 amplification patterns have been defined in tumors at different sites. Thus, amplification of cores 1 and 2 is less frequent in ovarian than breast cancer, in which all cores are usually amplified [24], [25], [26]. Moreover, CCND1 amplification has been reported in adenocarcinomas, melanoma, and squamous cell carcinomas [4], [21], [27], [28], whereas EMSY amplification appears to be frequent in breast adenocarcinoma but rare in squamous cell carcinomas [4], [29].

Section snippets

The 11q13 amplicon in OSCC

The 11q13 region is one of the most frequently amplified chromosome bands in head-and-neck squamous cell carcinoma (HNSCC), particularly in OSCC. Reported copy number gains of this chromosome band have ranged between 13 and 100% of OSCC cases [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48], [49] (Table 1), with a mean of 46%. However, higher amplification rates than those observed for the whole region have been reported for CCND1,

The 11q13 amplicon in oral potentially malignant disorders

Copy number gain or 11q13 chromosomal region amplification has been described in oral potentially malignant disorders (OPMDs) [41], [75], [76]. Cervigne et al. [75] found 11q13 band amplification in all degrees of epithelial dysplasia in leukoplakias that progressed to OSCC, while 11q13 gain was also reported by Noutomi et al. [41], mainly in the transition from moderate to severe epithelial dysplasia. Studies have been conducted on genes in the 11q13 amplicon that participate in early stages

CCND1

CCND1 encodes cyclin D1, which promotes cell cycle progression during G1 phase and favors G1-S transition [20]. The CCND1/cyclin D1 complex is the most widely studied component of chromosome band 11q13 in oral and other human cancers, including breast, lung, colon, and melanoma [85]. Besides its regulation of cell proliferation, numerous key functions in the cell biology of cancer have also been reported over recent years, including mitochondrial activity modulation, DNA repair, and cell

Candidate tumor suppressor genes in the 11q13 amplicon in OSCC

The PPFIA1 gene encodes Liprin-alpha-1, a member of the LAR protein-tyrosine phosphatase-interacting protein (liprin) family. Its functions are not well established but are thought to be related to the disassembly of focal adhesions at sites of cell-extracellular matrix interactions, with implications for cytoskeleton-matrix connections [111]. During the cascade of molecular phenomena and cellular interactions that enable metastasis of malignant epithelial cells, the cells must maintain an

The 11q13 amplicon as therapeutic target in OSCC

The CCND1/cyclin D1 complex is the most widely studied therapeutic target in 11q13 because there is greater knowledge of its clinicopathological implications. Cyclin D1 can be targeted in various ways: by acting on its cyclin dependent kinase (CDK) partners (CDK4/6), by acting directly on cyclin D1 or on the OSCC pathways in which cyclin D1 is involved, or by combining cyclin D1 inhibitors with other antitumor drugs [21], [114]. One promising therapeutic approach is to inhibit its CDK partners

Conclusions and final considerations

This review verifies the importance and complexity of 11q13 chromosome band amplification in OSCC. The co-amplification of a set of neighboring genes is frequent in this band, unlike other chromosomal regions, in which a single gene or a limited number of genes is typically amplified. To date, a single amplicon core has been confirmed in OSCC, located in chromosomal sub-region 11q13.2-q13.4; further studies are warranted to determine whether there is a second amplicon core in this tumor, as

Conflict of interest statement

None declared

References (129)

  • J.S. White et al.

    The influence of clinical and demographic risk factors on the establishment of head and neck squamous cell carcinoma cell lines

    Oral Oncol

    (2007)
  • K. Uchida et al.

    Molecular cytogenetic analysis of oral squamous cell carcinomas by comparative genomic hybridization, spectral karyotyping, and fluorescence in situ hybridization

    Cancer Genet Cytogenet

    (2006)
  • C. Garnis et al.

    OCGR array: an oral cancer genomic regional array for comparative genomic hybridization analysis

    Oral Oncol

    (2004)
  • E. Wolff et al.

    Oral squamous cell carcinomas are characterized by a rather uniform pattern of genomic imbalances detected by comparative genomic hybridisation

    Oral Oncol

    (1998)
  • L.Z. Luo et al.

    Cigarette smoke induces anaphase bridges and genomic imbalances in normal cells

    Mutat Res

    (2004)
  • C. Jin et al.

    Cytogenetic abnormalities in 106 oral squamous cell carcinomas

    Cancer Genet Cytogenet

    (2006)
  • K. Freier et al.

    Recurrent FGFR1 amplification and high FGFR1 protein expression in oral squamous cell carcinoma (OSCC)

    Oral Oncol

    (2007)
  • J. Xia et al.

    Amplifications of TAOS1 and EMS1 genes in oral carcinogenesis: association with clinicopathological features

    Oral Oncol

    (2007)
  • I. Wu et al.

    BNF-1, a novel gene encoding a putative extracellular matrix protein, is overexpressed in tumor tissues

    Gene

    (2003)
  • M.A. González-Moles et al.

    Β-catenin in oral cancer: an update on current knowledge

    Oral Oncol

    (2014)
  • H.-S. Liu et al.

    Detection of copy number amplification of cyclin D1 (CCND1) and cortactin (CTTN) in oral carcinoma and oral brushed samples from areca chewers

    Oral Oncol

    (2009)
  • J.C. de Vicente et al.

    Cortactin and focal adhesion kinase as predictors of cancer risk in patients with premalignant oral epithelial lesions

    Oral Oncol

    (2012)
  • X. Zhou et al.

    Global expression-based classification of lymph node metastasis and extracapsular spread of oral tongue squamous cell carcinoma

    Neoplasia

    (2006)
  • L. Tourneur et al.

    FADD: a regulator of life and death

    Trends Immunol

    (2010)
  • A.M. Chinnaiyan et al.

    FADD, a novel death domain-containing protein, interacts with the death domain of Fas and initiates apoptosis

    Cell

    (1995)
  • C. Lengauer et al.

    Genetic instabilities in human cancers

    Nature

    (1998)
  • D.G. Albertson et al.

    Chromosome aberrations in solid tumors

    Nat Genet

    (2003)
  • J. Tower

    Developmental gene amplification and origin regulation

    Annu Rev Genet

    (2004)
  • M. Schwab

    Amplification of oncogenes in human cancer cells

    BioEssays

    (1998)
  • P.M. Wilkerson et al.

    The 11q13-q14 amplicon: clinicopathological correlations and potential drivers

    Genes Chromosomes Cancer

    (2013)
  • R. Natrajan et al.

    Functional characterization of the 19q12 amplicon in grade III breast cancers

    Breast Cancer Res

    (2012)
  • H. Roelofs et al.

    Amplification of the 11q13 region in human carcinoma cell lines: a mechanistic view

    Genes Chromosomes Cancer

    (1993)
  • R. Lidereau et al.

    Amplification of the int-2 gene in primary human breast tumors

    Oncogene Res

    (1988)
  • D.J. Zhou et al.

    Amplification of human int-2 in breast cancers and squamous carcinomas

    Oncogene

    (1988)
  • J. Adelaide et al.

    Chromosomal localization of the hst oncogene and its co-amplification with the int.2 oncogene in a human melanoma

    Oncogene

    (1988)
  • T. Tsuda et al.

    High incidence of coamplification of hst-1 and int-2 genes in human esophageal carcinomas

    Cancer Res

    (1989)
  • T. Motokura et al.

    A novel cyclin encoded by a bcl1-linked candidate oncogene

    Nature

    (1991)
  • G.A. Lammie et al.

    D11S287, a putative oncogene on chromosome 11q13, is amplified and expressed in squamous cell and mammary carcinomas and linked to BCL-1

    Oncogene

    (1991)
  • C.J. Sherr et al.

    Living with or without cyclins and cyclin-dependent kinases

    Genes Dev

    (2004)
  • P. Ramos-García et al.

    An update on the implications of Cyclin D1 in oral carcinogenesis

    Oral Dis

    (2016)
  • C. Liang et al.

    Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG

    Nat Cell Biol

    (2006)
  • I. Dimova et al.

    Association of CyclinD1 copy number changes with histological type in ovarian tumors

    Acta Oncol

    (2004)
  • F. Courjal et al.

    Cyclin gene amplification and overexpression in breast and ovarian cancers: evidence for the selection of cyclin D1 in breast and cyclin E in ovarian tumors

    Int J Cancer

    (1996)
  • J.A. Curtin et al.

    Distinct sets of genetic alterations in melanoma

    N Engl J Med

    (2005)
  • D.M. Barnes et al.

    Cyclin D1 in breast cancer

    Breast Cancer Res Treat

    (1998)
  • P.M. Wilkerson et al.

    Functional characterization of EMSY gene amplification in human cancers

    J Pathol

    (2011)
  • P.M.W. van Kempen et al.

    Clinical relevance of copy number profiling in oral and oropharyngeal squamous cell carcinoma

    Cancer Med

    (2015)
  • K. Sugahara et al.

    Combination effects of distinct cores in 11q13 amplification region on cervical lymph node metastasis of oral squamous cell carcinoma

    Int J Oncol

    (2011)
  • K. Uchida et al.

    Loss of 3p26.3 is an independent prognostic factor in patients with oral squamous cell carcinoma

    Oncol Rep

    (2011)
  • S.M. Pathare et al.

    Clinicopathological and prognostic implications of genetic alterations in oral cancers

    Oncol Lett

    (2011)
  • Cited by (39)

    View all citing articles on Scopus
    View full text