Drug-induced increase of carcinoembryonic antigen expression in cancer cells

https://doi.org/10.1016/j.phrs.2003.12.007Get rights and content

Abstract

Most of gastrointestinal, breast and lung cancer cells express carcinoembryonic antigen (CEA). Therefore, this protein represents a suitable target for innovative diagnostic and immunotherapeutic strategies of various tumours. Presently CEA can be involved in three main approaches concerning cancer detection and therapy, i.e. (a) detection of tumour cells in the peripheral blood, bone marrow or lymph node using reverse transcriptase-polymerase chain reaction (RT-PCR)-based measurement of CEA mRNA; (b) targeting of anticancer agents or radionuclides by tumour-selective anti-CEA monoclonal antibodies (mAbs); (c) use of antitumour vaccines capable of eliciting major histocompatibility complex (MHC)-restricted immune responses against CEA-derived peptides.

Actually, it has been shown that the expression of CEA can be up-regulated by pharmacological agents including, antineoplastic drugs (i.e. 5-fluorouracil), cytokines (i.e. interferons or interleukin-6), differentiating agents (i.e. sodium butyrate) and protein kinase inhibitors (i.e. staurosporine). Therefore, the use of drugs capable of increasing CEA expression, could amplify the sensitivity of diagnostic procedures that rely on CEA determination. Moreover, the same agents could increase the efficacy of vaccines based on immunogenic CEA-derived peptides restricted by the MHC. The purpose of this review is to describe several agents that are able to increase CEA expression and to discuss the rational bases for new strategies in cancer detection and therapy aimed at increasing the expression of tumour-associated antigens.

Introduction

The carcinoembryonic antigen (CEA) is a glycoprotein containing approximately 50% of carbohydrate with a molecular weight of 180–200 kDa [1], [2], [3]. This protein belongs to the large CEA gene family, which includes 29 genes/pseudogenes [4], [5]. All genes of the CEA family are located within the long arm of chromosome 19 [6], [7]. CEA gene family can be divided into three subgroups, i.e. the CEA subgroup containing seven expressed genes; the pregnancy-specific-glycoprotein (PSG) containing 11 expressed genes and the third untitled subgroup containing six pseudogenes. Components of the CEA subgroup are cell surface bound proteins including CEA and the CEA cross reacting molecules, i.e. non-specific cross-reacting antigen (NCA), biliary glycoprotein (BGP) and CEA gene family member (CGM-6). As a tumour-associated antigen, CEA is extensively expressed in more than 95% of colorectal, gastric and pancreatic carcinomas as well as in approximately 70% of non-small cell lung cancer, 50% of breast cancer, in mucinous ovarian carcinoma, adenocarcinoma of the penis [8] and endometrial adenocarcinoma. CEA is also present, to a lesser extent, on normal adult human colon and in some foetal tissues [9], [10], [11], [12]. In particular, it is expressed on the mature columnar epithelial cells and goblet cells of colon, on pyloric mucous cells of stomach, on squamous epithelial cells of tongue, oesophagus and cervix, in secretory epithelia and duct cells of sweat glands and epithelial cells of the prostate [13].

The molecular basis for this differential expression in different tumours and normal tissues are not completely elucidated. The molecular cloning of cDNA for the CEA and CEA-related molecules was performed by different groups [14], [15], [16], [17], [18], [19], [20], [21]. Schrewe et al. [21] reported the isolation and characterisation of a cosmid clone that contains the complete coding region of the CEA gene, including its promoter sequence. No classic TATA or CAAT boxes have been found at the expected position upstream of the transcription sites of the CEA or NCA gene [21]. Approximately 400 bp upstream from the translation start within the promoter region confers cell-type-specific expression on a reported gene. Hauck and Stanners [22] also identified several nuclear factor binding sites such as upstream stimulatory factor (USF), Sp1, and Sp1-like factor, interacting with the promoter region of the CEA gene. CEA promoter region isolated from CEA-producing human colorectal carcinoma or normal adjacent mucosa are not qualitatively different [23]. It was further demonstrated that nuclear extracts from CEA-producing colorectal carcinoma cells could equally bind to both CEA promoter region isolated from colorectal carcinoma and from normal adjacent mucosa [23]. These studies suggest that DNA-binding CEA transcription factors are not sufficient to induce high levels of CEA expression in normal tissues. Therefore, some other mechanisms appear to be involved in the notable difference of CEA found in tumours compared with normal colonic mucosa. It has been demonstrated that methylation of CEA gene [24] and/or structural changes of transcription factors that interact with the regulatory elements [22] of the CEA promoter may increase CEA gene expression. Moreover, post-transcriptional modifications could also have a modulatory role.

The analysis of the amino acid sequences of the CEA family revealed that they belong to the immunoglobulin super-family [25]. CEA has been found to play a role as adhesion molecule with an important function in signal transduction in cancer cells. Several CEA subfamily members, including CEA, possess cell adhesion properties [26], [27], [28], [29], [30], and their presence in the tumour cell membrane has been considered responsible for increasing the tumour cell dissemination [31], [32], [33], [34], [35], [36].

Some authors suggested that CEA molecules expressed on normal cells might play a role in innate immune defence, protecting the colonic mucosa from microbial invasion [37]. Recently, Soeth et al. [38] reported that over expression of CEA protects human colon cancer cells from a variety of apoptotic stimuli, including drug treatment, UV light and confluent growth.

Identification of human tumour-associated antigens (TAAs) and epitopes as target for specific immunotherapy are currently under study. The TAAs that have been characterised in recent years, derive from a broad array of intracellular molecules such as point mutated ras [39], point mutated p53 [40] c-Erb/2, prostate-specific antigen [41], MUC-1 [42], and CEA [43], [44], [45]. It is well accepted that MHC-restricted peptides originated from these molecules are capable of being recognised by T cells. For example, CEA protein is processed endogenously and subsequently short peptide segments are presented by HLA alleles on tumour cell surface. Two distinct pathways have been identified for the processing of CEA as an immunogenic protein. In the first pathway, extra cellular antigen is processed for presentation as 15–25 amino acid length peptides to CD4+ T helper (TH) lymphocytes by MHC class II molecules found on the professional antigen-presenting cells (APCs) including dendritic cells and macrophages. In the second pathway, intracellular antigen produce processed peptides of eight or nine amino acids that are presented to CD8+ cytotoxic T lymphocytes (CTL) by MHC class I molecules (i.e. A2, A3, and A24).

These findings point out that CEA could represent a potential target molecule for specific immunotherapy including recombinant vaccines. A number of preclinical studies demonstrate the possibility to generate in vivo a CEA-specific CTL response with anti-tumour activity in mice and in non-human primates. These results have been obtained by using several strategies of immunisation such as recombinant viruses (CEA gene engineered poxoviruses, adenoviruses, etc.), recombinant DNA or anti-idiotypic antibodies [46], [47], [48], [49]. A human CTL-mediated response to CEA has been demonstrated by using CEA-derived peptides with HLA-A2.1 binding anchor motifs to stimulate in vitro human peripheral blood mononuclear cells (PBMC) derived from normal donors and patients with colorectal carcinoma [45]. As a consequence of such studies, several clinical trials are currently investigating the possible use of CEA as a target antigen, according to different protocols of active specific immunotherapy of patients with colo-rectal carcinoma [50], [51], [52]. In spite of encouraging preclinical results, the use of CEA-directed vaccines has been so far unsuccessful in controlling cancer progression in humans. One of the possible explanations is that CEA is heterogeneously expressed in the tumour. Therefore, it cannot be ruled out that a fraction of the entire tumour cell population would not express adequate amount of CEA peptide to be recognised by CEA-specific CTLs.

CEA is widely used as a tumour marker for diagnostic and therapeutic purposes in various neoplasias including gastrointestinal, breast and lung cancer. Since most of CEA positive colorectal tumours shed the antigen in the plasma, measurement of CEA levels is currently used to monitor tumour progression during the post surgery follow-up of patients affected by colorectal cancer. However, it should be noted that not all CEA positive tumours shed the antigen in the serum and no correlation has been found between CEA content in the tumour and CEA levels in the serum. Therefore, in a substantial number of cases this antigen cannot be considered a suitable marker for the detection of sub-clinical residual disease.

Previously, it has been demonstrated that methods based on reverse transcriptase-polymerase chain reaction (RT-PCR) for the detection of CEA positive circulating cancer cells, are sensitive and specific [53], [54], [55], [56]. However, limitations of this technique have also been reported in terms of false positive results when RNA was extracted from whole blood [57], [58]. Therefore, it has been proposed that epithelial tumour cell enrichment by immunomagnetic beads coated with an antibody raised against a common epithelial antigen would increase specificity of tumour-associated CEA transcript detection [59], [60], [61], [62]. Recently, real-time RT-PCR approach have been introduced as an alternative to conventional RT-PCR for sensitive and quantitative detection of CEA mRNA expressing micrometastasis in the peripheral blood [63], in lymph nodes [64] and in the peritoneal cavity [65] of gastroenteric cancer patients.

CEA has been also used for cancer cell targeting of antineoplastic agents or radionuclides bound to tumour-selective anti-CEA monoclonal antibodies (mAbs) [66]. This is an attractive approach either for therapeutic or diagnostic purpose. In fact, anti-CEA mAbs conjugated with 123I have been demonstrated to be safe and effective in tumour localisation in cancer patients [67]. Additional studies have shown that 131I radiolabelled mAbs provide promising therapeutic effects in terms of reduction of tumour mass and metastasis [68], [69], [70], [71], [72]. Moreover, CEA expression might be involved in radioimmunoguided surgery for the recognition of tumour cells by anti-CEA radiolabelled mAbs [73], [74].

Important factors that appear to limit the use of tumour markers for antibody-based diagnosis and therapy are the low expression of target molecules, the poor penetration of whole antibodies and the formation of human anti-mouse antibodies against to the administered antibody. Therefore, agents capable of increasing CEA expression might represent suitable tools to improve sensitivity and efficacy of diagnostic and therapeutic approaches based on this tumour marker. In particular, pharmacological increment of tumour-associated CEA could be accompanied by higher chances of presenting MHC-restricted CEA-derived peptides by cancer cells. It follows that drug-mediated increase of CEA expression could facilitate the recognition of target cells by cell-mediated immune response induced by anti-CEA peptide vaccines. It was also demonstrated that agents which increase CEA expression can be used to improve either the antitumour effect of radionuclide-conjugated anti-CEA mAbs [72], [75], [76] or the immunodiagnostic procedures [77] such as radioimmunoscintigraphy or radioimmunoguided surgery based on recognition of this marker. Moreover, drug-induced enhancement of CEA might improve sensitivity of RT-PCR-based detection of circulating tumour cells from peripheral blood of cancer patients [61], [62].

A summary of the agents alone or in combination reviewed in this article and their effects on CEA expression/secretion is shown in Table 1, Table 2, respectively.

Section snippets

Interferons

Attallah et al. [78] observed for the first time that up-regulation of CEA could be obtained in a human colon carcinoma cell line, WiDr, after treatment with a preparation of lymphoblastoid interferon. Further studies have compared the ability of three different human interferons (IFN-α, -β, -γ) to increase the expression of tumour-associated antigen, including CEA, on freshly-isolated human adenocarcinoma cells [79] and on established cancer cell lines [80], [81]. The results revealed that

Conclusion

CEA is present in the digestive organs of the human foetus. However, the majority of gastrointestinal cancer as well as breast cancer and small cell lung carcinoma express CEA. A growing body of literature established that the measure of CEA levels in the plasma could not be considered a suitable marker for the screening of colon cancer [132]. However, there is good evidence that the routine serum CEA determination in post-surgical surveillance of colon cancer reveals early recurrence.

A number

Acknowledgements

The work in Dr. A. Aquino’s laboratory was supported by a grant from “Istituto Superiore di Sanità” n.1A2/F7 (Research unit, Enzo Bonmassar).

References (159)

  • B. Obrink

    CEA adhesion molecules: multifunctional proteins with signal-regulatory properties

    Curr. Opin. Cell. Biol.

    (1997)
  • S. Kuninaka et al.

    Direct influences of pro-inflammatory cytokines (IL-1beta, TNF-alpha, IL-6) on the proliferation and cell-surface antigen expression of cancer cells

    Cytokine

    (2000)
  • S. Hammarstrom et al.

    Is there a role for CEA in innate immunity in the colon?

    Trends Microbiol.

    (2001)
  • J.A. Bristol et al.

    Identification of a ras oncogene peptide that contains both CD4(+) and CD8(+) T cell epitopes in a nested configuration and elicits both T cell subset responses by peptide or DNA immunization

    Cell. Immunol.

    (2000)
  • S. Ito et al.

    Quantitative detection of CEA expressing free tumor cells in the peripheral blood of colorectal cancer patients during surgery with real-time RT-PCR on a LightCycler

    Cancer Lett.

    (2002)
  • G. Hajjar et al.

    Phase I radioimmunotherapy trial with iodine-131-labeled humanized MN-14 anti-carcinoembryonic antigen monoclonal antibody in patients with metastatic gastrointestinal and colorectal cancer

    Clin. Colorectal Cancer

    (2002)
  • T. Yano et al.

    Direct influence of interferon-gamma on proliferation and cell-surface antigen expression of non-small cell lung cancer cells

    Cancer

    (2000)
  • S. Hammarstrom et al.

    Nature of the tumor-associated determinant(s) of carcinoembryonic antigen

    Proc. Natl. Acad. Sci. U.S.A.

    (1975)
  • W. Zimmermann et al.

    Chromosomal localization of the carcinoembryonic antigen gene family and differential expression in various tumors

    Cancer Res.

    (1988)
  • J. Thompson et al.

    Long-range chromosomal mapping of the carcinoembryonic antigen (CEA) gene family cluster

    Genomics

    (1992)
  • A.L. Cubilla et al.

    Surface adenosquamous carcinoma of the penis. A report of three cases

    Am. J. Surg. Pathol.

    (1996)
  • F. Prall et al.

    CD66a (BGP), an adhesion molecule of the carcinoembryonic antigen family, is expressed in epithelium, endothelium, and myeloid cells in a wide range of normal human tissues

    J. Histochem. Cytochem.

    (1996)
  • M. Nap et al.

    Immunohistochemistry of carcino-embryonic antigen in the embryo, fetus and adult

    Tumor Biol.

    (1988)
  • M. Nap et al.

    Specificity and affinity of monoclonal antibodies against carcinoembryonic antigen

    Cancer Res.

    (1992)
  • P.F. Robbins et al.

    Definition of the expression of the human carcinoembryonic antigen and non-specific cross-reacting antigen in human breast and lung carcinomas

    Int. J. Cancer

    (1993)
  • L. Frangsmyr et al.

    Four carcinoembryonic antigen subfamily members, CEA, NCA, BGP and CGM2, selectively expressed in the normal human colonic epithelium, are integral components of the fuzzy coat

    Tumor Biol.

    (1999)
  • N. Beauchemin et al.

    Isolation and characterization of full-length functional cDNA clones for human carcinoembryonic antigen

    Mol. Cell. Biol.

    (1987)
  • M. Neumaier et al.

    Characterization of a cDNA clone for the nonspecific cross-reacting antigen (NCA) and a comparison of NCA and carcinoembryonic antigen

    J. Biol. Chem.

    (1988)
  • Y. Hinoda et al.

    Molecular cloning of a cDNA coding biliary glycoprotein I: primary structure of a glycoprotein immunologically crossreactive with carcinoembryonic antigen

    Proc. Natl. Acad. Sci. U.S.A.

    (1988)
  • B. Berling et al.

    Cloning of a carcinoembryonic antigen gene family member expressed in leukocytes of chronic myeloid leukemia patients and bone marrow

    Cancer Res.

    (1990)
  • H. Schrewe et al.

    Cloning of the complete gene for carcinoembryonic antigen: analysis of its promoter indicates a region conveying cell type-specific expression

    Mol. Cell. Biol.

    (1990)
  • G. Cao et al.

    Comparison of carcinoembryonic antigen promoter regions isolated from human colorectal carcinoma and normal adjacent mucosa to induce strong tumor-selective gene expression

    Int. J. Cancer

    (1998)
  • R. Tran et al.

    Correlation of DNA hypomethylation with expression of carcinoembryonic antigen in human colon carcinoma cells

    Cancer Res.

    (1988)
  • R.J. Paxton et al.

    Sequence analysis of carcinoembryonic antigen: identification of glycosylation sites and homology with the immunoglobulin supergene family

    Proc. Natl. Acad. Sci. U.S.A.

    (1987)
  • J.M. Jessup et al.

    Adhesion to carcinoembryonic antigen by human colorectal carcinoma cells involves at least two epitopes

    Int. J. Cancer

    (1993)
  • H. Zhou et al.

    Specificity of anti-carcinoembryonic antigen monoclonal antibodies and their effects on CEA-mediated adhesion

    Cancer Res.

    (1993)
  • J.M. Jessup et al.

    Carcinoembryonic antigen: function in metastasis by human colorectal carcinoma

    Cancer Metastasis Rev.

    (1989)
  • R.B. Hostetter et al.

    Carcinoembryonic antigen as a selective enhancer of colorectal cancer metastasis

    J. Natl. Cancer Inst.

    (1990)
  • D.W. Ohannesian et al.

    Carcinoembryonic antigen and other glycoconjugates act as ligands for galectin-3 in human colon carcinoma cells

    Cancer Res.

    (1995)
  • R. Zimmer et al.

    Expression profiling and interferon-beta regulation of liver metastases in colorectal cancer cells

    Clin. Exp. Metastasis

    (2002)
  • T. Wirth et al.

    Inhibition of endogenous carcinoembryonic antigen (CEA) increases the apoptotic rate of colon cancer cells and inhibits metastatic tumor growth

    Clin. Exp. Metastasis

    (2002)
  • E. Soeth et al.

    Controlled ribozyme targeting demonstrates an antiapoptotic effect of carcinoembryonic antigen in HT29 colon cancer cells

    Clin. Cancer Res.

    (2001)
  • T.R. Petersen et al.

    Identification and design of p53-derived HLA-A2-binding peptides with increased CTL immunogenicity

    Scand. J. Immunol.

    (2001)
  • A. Heiser et al.

    Autologous dendritic cells transfected with prostate-specific antigen RNA stimulate CTL responses against metastatic prostate tumors

    J. Clin. Invest.

    (2002)
  • R.A. Henderson et al.

    Retroviral expression of MUC-1 human tumor antigen with intact repeat structure and capacity to elicit immunity in vivo

    J. Immunother.

    (1998)
  • R.M. Conry et al.

    Immune response to a carcinoembryonic antigen polynucleotide vaccine

    Cancer Res.

    (1994)
  • S.K. Nair et al.

    Induction of primary carcinoembryonic antigen (CEA)-specific cytotoxic T lymphocytes in vitro using human dendritic cells transfected with RNA

    Nat. Biotechnol.

    (1998)
  • K.Y. Tsang et al.

    Generation of human cytotoxic T cells specific for human carcinoembryonic antigen epitopes from patients immunized with recombinant vaccinia-CEA vaccine

    J. Natl. Cancer Inst.

    (1995)
  • M.Z. Zhu et al.

    Specific cytolytic T-cell responses to human CEA from patients immunized with recombinant avipox-CEA vaccine

    Clin. Cancer Res.

    (2000)
  • K.A. Foon et al.

    Clinical and immune responses in advanced colorectal cancer patients treated with anti-idiotype monoclonal antibody vaccine that mimics the carcinoembryonic antigen

    Clin. Cancer Res.

    (1997)
  • Cited by (57)

    • A novel sandwich-type SERS immunosensor for selective and sensitive carcinoembryonic antigen (CEA) detection

      2020, Analytica Chimica Acta
      Citation Excerpt :

      It was first discovered by Gold and Freedman in 1965 and was initially thought to be specific to colon cancer. Then, It was characterized immunologically by selective adsorption and was found to be a glycoprotein family [3,4]. It is not only specific to the colon but also increases in other lung, pancreas and prostate cancers.

    • Fabrication of sensitive bioelectrode based on atomically thin CVD grown graphene for cancer biomarker detection

      2018, Biosensors and Bioelectronics
      Citation Excerpt :

      The early and accurate detection of cancer is extremely important for clinical diagnosis, effective toxicity monitoring, and ultimately for the successful treatment of cancer. CEA is one of the cancer biomarkers, which has been extensively used for diagnostic purposes in gastrointestinal, breast and lung cancer (Aquino et al., 2004; Duffy, 2001). In an adult nonsmoker human serum, the normal level of CEA is less than 2.5 ng mL−1 and for a smoker it is less than 5.0 ng mL−1 (Loewenstein and Zamcheck, 1978; Su et al., 2008).

    • Tunicamycin impairs olfactory learning and synaptic plasticity in the olfactory bulb

      2017, Neuroscience
      Citation Excerpt :

      Previous studies have utilized tunicamycin (TM) as a toxin to reveal the pathology of various diseases or develop therapeutic agents (Elbein, 1984; Schwarz, 1991; Aquino et al., 2004).

    View all citing articles on Scopus
    View full text