Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Membrane-tethered mucin glycoproteins are abundantly expressed at the apical surfaces of simple epithelia, where they play important roles in lubricating and protecting tissues from pathogens and enzymatic attack. Notable examples of these mucins are MUC1, MUC4 and MUC16 (also known as cancer antigen 125). In adenocarcinomas, apical mucin restriction is lost and overall expression is often highly increased. High-level mucin expression protects tumors from killing by the host immune system, as well as by chemotherapeutic agents, and affords protection from apoptosis. Mucin expression can increase as the result of gene duplication and/or in response to hormones, cytokines and growth factors prevalent in the tumor milieu. Rises in the normally low levels of mucin fragments in serum have been used as markers of disease, such as tumor burden, for many years. Currently, several approaches are being examined that target mucins for immunization or nanomedicine using mucin-specific antibodies.

Free full text 


Logo of nihpaLink to Publisher's site
Expert Rev Endocrinol Metab. Author manuscript; available in PMC 2012 Sep 1.
Published in final edited form as:
Expert Rev Endocrinol Metab. 2011 Nov; 6(6): 835–848.
https://doi.org/10.1586/eem.11.70
PMCID: PMC3245640
NIHMSID: NIHMS340062
PMID: 22201009

Transmembrane mucins as novel therapeutic targets

Abstract

Membrane-tethered mucin glycoproteins are abundantly expressed at the apical surfaces of simple epithelia, where they play important roles in lubricating and protecting tissues from pathogens and enzymatic attack. Notable examples of these mucins are MUC1, MUC4 and MUC16 (also known as cancer antigen 125). In adenocarcinomas, apical mucin restriction is lost and overall expression is often highly increased. High-level mucin expression protects tumors from killing by the host immune system, as well as by chemotherapeutic agents, and affords protection from apoptosis. Mucin expression can increase as the result of gene duplication and/or in response to hormones, cytokines and growth factors prevalent in the tumor milieu. Rises in the normally low levels of mucin fragments in serum have been used as markers of disease, such as tumor burden, for many years. Currently, several approaches are being examined that target mucins for immunization or nanomedicine using mucin-specific antibodies.

Keywords: cytokines, MUC1, MUC16, MUC4, nanomedicine, steroid hormones

Apical mucosal surfaces are dominated by a thick forest of high-molecular-weight, heavily glycosylated proteins collectively referred to as mucins. Mucin glycoproteins are characterized as proteins heavily substituted with O-linked oligosaccharides, most often in tandem repeat domains rich in serine, threonine and proline residues. The resulting molecules are extremely hydrophilic with physically extended structures supported by the large hydration sphere of the oligosaccharides as well as the structural rigidity conferred by proline residues. At least 21 mucin genes exist in humans [1]. Mucins can be subdivided into two classes: secreted or gel-forming, and cell surface or trans-membrane [2]. The strict classification of the latter is complicated by the occurrence of splice variants that generate secreted forms [3,4], as well as post-translational processing events that cleave the protein core, separating the mucin domains from their membrane anchors [59]. Nonetheless, it appears that the membrane-anchored forms of these gene products are the predominant forms in most situations. The current article will focus on three complex, transmembrane mucins, MUC1, MUC4 and MUC16. The unique features and functions of each mucin will be discussed, as well as the opportunities these molecules present as novel therapeutic targets.

Mucin structure & synthesis

The generic structures of transmembrane mucins consist of a very large extracellular domain dominated by tandem repeat motifs of sequences that function as acceptors of O-linked oligosaccharides, a single, short transmembrane domain and a relatively small cytoplasmic tail. Furthermore, as discussed later, the MUC4 and MUC16 extracellular domains have a number of other structural features. Mucin mRNAs encode N-terminal signal sequences and are translated in the rough endoplasmic reticulum where core N-glycosylation occurs; however, most of the glycosylation reactions of mucin-type oligosaccharides occur in the Golgi apparatus [10,11]. Mucin-type oligosaccharides vary in size, being as small as one saccharide unit (approximately 200 MW) or substantially larger (Figure 1C). Mucin ectodomains typically contain hundreds of these oligosaccharides, giving them a very large and heterogeneous size distribution. The oligosaccharides hydrate and contribute to the lubricating and generally anti-adhesive functions of these molecules. Nonetheless, certain sialic acid and sulfation modifications can occur that generate siglec and selectin ligands, thereby creating adhesion-promoting motifs [12,13]. The oligosaccharides may also serve as binding sites for bacteria [14,15]. It is not clear if different mucin glycoproteins expressed by the same cell express distinct profiles of mucin oligosaccharides; however, there is no evidence to indicate that the tandem motifs present in different mucin core proteins are differentially glycosylated. The glycomic analyses done to date focus on total glycan profiles from a given mucin or mucin fragment. It remains possible that glycan structural differences occur at specific sites or in response to physiologic stimuli, such as hormones or cytokines.

An external file that holds a picture, illustration, etc.
Object name is nihms340062f1.jpg
Membrane-bound mucins

(A) The sizes of typical cell surface receptors, such as intergrins and EGFR, relative to those of MUC1, MUC4 and MUC16 are shown. (B) Domain organization of MUC1, MUC4 and MUC16. (C) Cancer-associated antigens (Tn, STn and T/TF) result from aberrant O-glycosylation on the ectodomains of MUC1, 4 and 16. O-linked glycosylation is initiated by adding GalNAc to the protein backbone, resulting in the Tn antigen. NeuAc of the GalNAc moiety results in the formation of the STn antigen. Alternatively, core 1 structures, T and TF, are synthesized by the addition of Gal to the Tn antigen. Non-cancer-associated antigens are formed from either the sialylation of the core 1 structures, resulting in the mono- or disialyl-T antigen, or the continued branching/elongation by adding GlcNAc to form the core 2 structures. Further elongation of the core 2 structure forms more complex peripheral antigens, such as the variations of Lewis and Sialyl–Lewis. (D) O-glycosylation occurs on either a Ser or Thr residue within the tandem repeat sequences of the mucin ectodomains. The consensus sequences for the tandem repeats in human MUC1, MUC4 and MUC16 are shown using the single letter amino acid code. Serine and threonine residues are underlined. Note the large number of proline residues in these motifs.

AMOP: Adhesion-associated domain; CT: Cytoplasmic tail; CYS rich: Cysteine-rich domain; EGF repeats: EGF-like repeat region; EGFR: EGF receptor; Gal: Galactose; GalNAc: N-acetylgalactosamine; GlcNAc: N-acetylglucosamine; MUC: Mucin; NeuAc: Sialylation; NIDO: Nidogen-like domain; SEA: Sea urchin sperm receptor enterokinase-agrin domain; Ser: Serine; ST: Disialyl-T; STn: Sialyl-Tn; T/TF: Thomsen–Friedenreich antigen; Thr: Threonine; TM: Transmembrane domain; Tn: Precursor to T antigen; VWD: von Willebrand factor type D domain.

Excellent, detailed reviews on the membrane-anchored mucins are available [2,16]. A comparison of the relative sizes of mucin core proteins and major structural features is shown in Figures 1A & 1B. The majority of MUC1’s extracellular domain consists of a series of 20–21-amino-acid tandem repeats. In humans, the number of these repeats varies (25–125) in different MUC1 alleles, accounting for allelic polymorphism and different sizes of the mature glycoproteins ranging from 250 kDA to >1 MDa [17]. In addition, splice variants and differential glycosylation contribute to polymorphisms in mature MUC1 [18]. Other regions of the ecto-domain are also heavily O-glycosylated. Near the transmembrane region is a sperm protein, enterokinase and agrin domain containing an autocleavage site. Although this autocleavage appears to be quite efficient, the two resulting MUC1 subunits remain tightly associated [5,6]. MUC1’s C-terminal region consists of a highly conserved 72-amino-acid cytoplasmic tail region with many proposed functions (see Table 1 and discussion later).

Table 1

Membrane-bound mucins.

FunctionMUC1MUC4MUC16Ref.
Ectodomain
Anti-adhesive++?[2021]
Proadhesive+?+[22,2425]
Lubrication+++Reviewed in [2]
ERBB signaling++?Reviewed in [16,26,38]

Cytoplasmic tail
Phosphorylation sites+PotentialPotentialReviewed in
Signal transduction+??[16,43]
Transcriptional regulation+??

+: Known to be present; ?: Unknown.

MUC4 is substantially larger and more complex than MUC1, consisting of a complex of two subunits generated from a single polypeptide precursor [7]. The N-terminal α subunit ranges in size from 3000 to 7300 amino acids and contains a series of imperfect repeats, an O-glycosylated region of 145–395 variable number tandem 16 amino acid repeats, a unique sequence, a cysteine-rich domain, a nidogen homology domain (NIDO), an adhesion-associated domain (AMOP) and a von Willebrand factor D sequence (VWD) prior to its proteolytic cleavage site. The C-terminal β subunit following the proteolytic cleavage site contains 1156 amino acids and consists of an N-glycosylated region containing three EGF-like domains, a single transmembrane spanning sequence and a 22-amino-acid cytoplasmic tail.

MUC16 is by far the largest of the group, with an overall core protein size of approximately 22,000 amino acids (ca. 2.5 MDa). The N-terminal region consists of a very large, heavily O-glycosylated region, a region of more than 60–156 amino acid tandem repeats carrying both N- and O-linked oligosaccharides, and a membrane proximal region containing 16 sperm protein, enterokinase and agrin domains. Characterization of the N- and O-linked oligo-saccharides associated with MUC16 (cancer antigen [CA] 125) isolated from an ovarian cancer cell line has been reported, with both classes containing some interesting structural features [19]. The much smaller C-terminal region contains a single transmembrane region followed by a 32-amino-acid cytoplasmic tail.

Functions

A major, shared function of the ectodomains of these membrane-tethered mucins is to hydrate and lubricate cell surfaces. The ecto-domains of MUC1 and MUC4 are also anti-adhesive, a function that requires a critical, minimal number of tandem repeats to afford sufficient steric hindrance to cell surface receptors [20,21]. As mentioned previously, MUC1 can also carry selectin ligands and, therefore, may support cell attachment in some instances [22]. An anti-adhesive function for MUC16 also seems likely since like MUC1, MUC16 inhibits synapse formation between natural killer cells and their targets [23]. Interestingly, MUC16 is proposed to bind both Siglec-9 [24] and mesothelin [25] and, therefore, may support cell attachment in certain scenarios. Other functions are presented in Table 1 and the discussion after.

MUC1 and MUC4 also complex with members of the ERBB family of receptors and modulate signal transduction mediated by these receptors (reviewed in [16,26]). In MUC1, this interaction requires the presence of the cytoplasmic tail, which becomes tyro-sine phosphorylated upon ERBB ligand addition. In MUC4, the interaction appears to involve its EGF-like domains acting like chaperones as well as weak receptor ligands. The cytoplasmic tail of MUC1 is also involved in a large array of other signal transduc-tion events including modulation of gene transcription, complexing with various cytoplasmic signaling proteins and protection of cells from apoptosis (Table 1; reviewed in [27]).

Interesting, yet still poorly understood, are the effects of trans-membrane mucins and their shed fragments on the immune system. Vasir et al. demonstrated that MUC1 is not only induced in T cells by IL-7, but also polarizes at the site of T cell–dendritic cell synapses [28]. In other studies, these mucins inhibit immune recognition [2830], apparently by sterically inhibiting access to the cell surface, and/or can be immunosuppressive [31,32]. Thus, these mucins contribute to complex, protective functions with regard to the immune system that, in the context of tumors, can protect the tumor cells from host immune surveillance.

MUC1-null mice have been created with phenotypes including slower growth of T-antigen-induced mammary tumors and increased susceptibility to infection/inflammation [33,34], although at least some of the latter responses are dependent on the genetic background of the mice used [35]. MUC16-null mice were recently created with no obvious phenotype, at least with regard to fertility and embryo/fetal development [36]. Interestingly, some reduction in MUC1 mRNA was evident in MUC16-null uteri, indicating that increased expression of MUC1 did not compensate for loss of MUC16. Mice double null for MUC1 and MUC16 have not been reported, and neither have MUC4-null mice.

Regulation of mucin expression

Membrane-tethered mucins demonstrate a tissue-specific distribution, suggesting a controlled regulation of their expression. Most epithelial cells produce mucins, and relative amounts of MUC1, MUC4 and MUC16 may vary depending on cell and tissue type [16,37].

Tissue distribution

MUC1 is expressed on the apical surface of nearly all simple epithelial tissues, as well as tumor cells. MUC1 expression is associated with epithelial cell differentiation in the stomach, pancreas, lung, trachea, kidney, salivary and mammary glands and the female reproductive tract [16,38,39]. MUC1 is also expressed on certain nonepithelial cells such as hematopoietic cells, activated T cells and sperm [4043]. MUC4 is also expressed in many normal epithelial tissues, both during development and in adults, including those of the eye, oral cavity, lacrimal glands, salivary gland, female reproductive tract, prostate gland, stomach, colon, lung, trachea and mammary gland. In lungs, MUC4 is the first mucin to be expressed [16,44]. MUC16, widely known as the serum marker of ovarian cancer (CA 125), is expressed on the epithelial cells of the eye, respiratory tract, female reproductive tract and the mesothelium of the abdominal cavity [4548]. Of the three membrane mucins discussed, MUC16 is the least well characterized. While a variety of monoclonal antibodies to MUC16 (CA 125) have been described, their reactivities differ significantly [49] and may be influenced by glycosylation.

MUC1, MUC4 and MUC16 expression in cancer progression and metastasis is characterized by increased levels, altered gly-cosylation and aberrant surface distribution patterns. Aberrant expression of MUC1 in several adenocarcinomas, including breast, pancreas, colon, lung and endometrial cancer, is well established. However, in the case of prostate cancer, MUC1 expression is a poor marker of progression [50]. MUC4 is aberrantly expressed in ovarian tumors and premalignant and malignant pancreatic lesions. In prostate carcinomas, MUC4 expression is significantly downregulated [51]. MUC16 expression has been well investigated in ovarian carcinoma, and it is highly expressed in both ovarian and endometrial cancers [52,53]. A more detailed discussion on mucins in cancer is provided in the following sections.

Transcriptional regulation

Understanding the molecular controls over mucin gene expression may provide new opportunities to therapeutically intervene to manipulate mucin levels to enhance or reduce their function. Transcriptional regulation of MUC1 and MUC4 has been reviewed in detail [44,54]. Based on the epithelial-specific expression of human MUC1 in transgenic mice, it was determined that the regulatory elements necessary for normal patterns are within the proximal 1.4-kb region of the MUC1 promoter. The highly active region of the MUC1 promoter is encompassed within the −600 +1 base pair region of the transcriptional start site and has multiple response elements including those for Sp1, an E-box, signal transducers and activators of transcription (STATs) and NF-κB family members, a P-responsive region (PRE) and peroxisome proliferator-activated receptor (PPAR) responsive region [54]. Proinflammatory cytokines greatly stimulate MUC1 expression in various cellular contexts [54]. Estrogen (E2) and progesterone (P) regulate mucin gene expression in the female reproductive tract of humans and rodents. Steroid hormonal influences on MUC1 gene expression has been reviewed in detail by Carson et al. [54]. Rodent MUC4 expression increases with high levels of E2 and decreases when P levels are dominant [55,56]. Similar to murine MUC1 regulation, E2 stimulates rodent MUC4 expression [56,57]. MUC4 expression is also reduced at time of implantation [58] in rats. Both MUC4 and MUC16 expression have been profiled during the menstrual cycle; however, intensive studies to understand regulation by steroid hormones are yet to be investigated [59,60]. Loss of MUC16 also may aid blasctocyst adherence [60]. In addition, P receptor (PR) regulates human MUC1 gene expression in a PR isoform-specific fashion [61]. Progesterone-stimulated MUC1 expression is antagonized by PPAR-γ [62] in both uterine epithelial and cancer cells. MUC1 gene expression is also modulated by a dynamic interplay among cytokine-activated transcription factors, PR isoforms and transcriptional coregulators such as p300 and SRC-3 [63].

Characterization of the proximal 3.7 kb of the MUC4 5′-flank-ing region revealed four transcriptional start sites, three in the distal region and one in the proximal region. The proximal region is TATA-less, while the distal promoter is characterized by a TATA box [64]. Numerous transcription factor binding sites important for initiation of transcription are located within the proximal promoter. Transcriptional regulation of MUC4 has been examined in some detail in pancreatic cancer cells. IFN-γ activates MUC4 expression via STAT-1 upregulation [65]. TGF-β also activates MUC4 expression [66]. Retinoic acid (RA) increases MUC4 expression via RAR-α [67]. IFN-γ and RA synergize at the transcriptional level to induce MUC4 expression in pancreatic cancer cells [68]. IFN-γ and TNF-α activate MUC4 transcription via STAT and NF-κB family members [64]. Interleukins (IL-4 and IL-9) also regulate MUC4 transcription. Other transcription factors that regulate MUC4 expression are hepatocyte nuclear factors (HNF-1/-4), forkhead box A (FOX A1/A2), GATA (-4/-5/-6) and caudal-related homeobox (CDX -1/-2) [69,70].

In human corneal epithelial cells, IFN-γ and TNF-α modulate MUC16 protein expression and ectodomain release [71]. Nonetheless, regulation of MUC16 gene expression and functional analyses of the MUC16 promoter have yet to be examined. Putative binding sites for transcription factors, such as ER, STAT1, STAT3, CRE-BP1 (ATF2) and NF-κB, are evident by sequence inspection [72]; however, functional studies have not been performed to determine a true role for these elements. Recent developments have been made in understanding epigenetic control of mucin transcription in cancer, as reviewed by Van Seuningen and Vincent, for enabling new therapeutic approaches [73].

Post-transcriptional regulation

The full-length splice variant of MUC1 is also known as MUC1/REP [74]. Alternate forms of MUC1 mRNA give rise to MUC1 isoforms lacking the transmembrane and cytoplasmic domains, MUC1/SEC [74] or the tandem repeat region, MUC1/Y and MUC1/X [75,76]. MUC1/X was also reported as MUC1/Z [77]. Another splice variant of MUC1 is MUC1/ZD, which lacks the tandem repeat leading to a frameshift in the cytoplasmic tail sequence [78]. Although these truncated forms are coexpressed with the full length MUC1, the full-length form is predominantly expressed.

Alternative splicing of the MUC4 transcript generates a large series of splice variants [79]. A total of 24 different MUC4 splice variants (sv0 [full length] to sv21-MUC4; MUC4X and MUC4/Y) have been categorized into three distinct families: a secreted family with 12 variants, a membrane-bound family with five variants and others that include the membrane-bound form, but lack the tandem repeat domain (MUC4/X and MUC4/Y) [3,79,80]. MUC4 splice variants have been studied primarily in pancreatic carcinoma cell lines; however, their expression is not detected in the normal pancreas [3,79,80]. The functional significance of these mRNA splice variants is unknown because of the lack of corresponding protein expression data. Although CA 125, the secreted form of MUC16, has been widely accepted as a marker for ovarian cancer, little is known regarding its transcriptional or post-transcriptional regulation.

Mucins in cancer

Both secreted and membrane-bound mucins are well established as tumor markers [81,82], as they are commonly overexpressed, underglycosylated [2,83,84] and associated with a poor prognosis in many carcinomas [8594]. Their barrier function potentially creates a local microenvironment, physically and chemically protecting metastatic cells from possible unfavorable conditions during invasion and tumor growth. The steric and charged properties of this barrier can facilitate or inhibit molecular diffusion, increasing the capture of growth factors or cytokines that contribute to tumor growth, protect tumor cells from immune system attacks, and inhibit uptake of many hydrophobic chemotherapeutic drugs [2,95,96]. Moreover, this barrier also reduces cell–cell and cell–extracellular matrix adhesion through both steric hindrance and by binding and occupying adhesion molecules [97,98], further promoting metastatic activity [2,28,99101]. In tumor cells, membrane-bound mucins lose their normal restriction to the apical cell surface and instead are found distributed over the entire cell surface, allowing interactions with growth factor receptors normally sequestered to the basolateral membrane. MUC1’s cytoplasmic tail also participates in multiple signal transduction processes in cancer cells (Table 1).

Unlike MUC1, MUC4 contains an EGF-like motif within its juxtamembrane ectodomain and is capable of interacting with ErbB2 to enhance oncogenic pathways in cancer cells [2]. In polarized cells expressing MUC4, this complex segregates ErbB2 in the apical cell membrane, away from ErbB3 in the lateral membrane. However, when polarity is lost, these two ErbB family members are permitted to interact, driving cell proliferation, motility and inhibition of apoptosis [102]. Similar to MUC1, MUC4 inactivates proapoptotic Bad and stabilizes anti-apoptotic Bcl-xL [103].

The biological significance of MUC16 overexpression in many cancers is poorly understood and studies have mostly focused on clinical applications. Although the MUC16 cytoplasmic domain contains potential phosphorylation sites [16], there is minimal evidence of cell signaling involvement. Recently, MUC16 demonstrated a possible role in ovarian cancer cell epithelial–mesen-chymal transition, presumably through the modulation of EGFR phosphorylation [104]. Extracellularly, MUC16 has been shown to contribute to ovarian cancer metastasis [25,105] and host immune response suppression [106].

Aberrant glycosylation of mucins in cancer

The characteristic patterns of reduced O-linked glycosylation expose and alter the conformation of the variable number tandem repeats (VNTRs) within the ectodomains of mucins. The exposed cancer-related epitopes can be tissue- and cancer-grade-specific. Altered glycosyltransferase expression in cancer cells leads to truncated O-linked glycan structures and high mannose and bisecting type N-linked glycans. This aberrant glycosylation can form several well-known core oligosaccharide antigens associated with tumors – Tn, sialyl-Tn (STn), Thomsen–Friedenreich (T or TF) and sialyl-T (ST) (Figure 1C). Moreover, complex peripheral O-linked oligosaccharide antigens can also form from further glycosylation of the core 2 antigen, for example, the variations of Lewis and Sialyl–Lewis antigens [19,107109]. Clinicians and researchers have taken advantage of these antigens, using monoclonal antibodies specific towards these unique carbohydrate targets and exposed protein core to distinguish cancerous from healthy tissue [36,110112]. Interestingly, the overexpression of sialyltransferases and subsequential increased sialylation is in itself believed to be important in the progression of many types of cancer [113120]; however, a mechanism has not been proposed.

Cancer diagnosis & prognosis

Abnormal mucin expression and/or glycosylation has been observed in the sera and tumors of various cancers, including lung, colon, pancreatic, ovarian and breast [109,121,122]. Heterogeneous mucin expression within types of cancer overlapping patterns among different types of cancer, and increased serum levels observed in benign conditions limit their use as diagnostic markers in determining the primary sites of metastatic disease [123].

An example of this is seen in breast cancer, where overexpression of all three of these mucins has been observed [85,121,122,124]. Although increased serum MUC1 in breast cancer patients is associated with higher tumor burden [125,126], no correlation has been shown towards tumorigenicity or metastasis [127]. However, increased levels of aberrantly glycosylated MUC1 has been shown to be a predictor of poor chemotherapeutic response [128] and an almost twice as low 5-year survival rate [129]. In addition, an accumulation of cytoplasmic MUC1 is also associated with a lower breast cancer survival rate [85]. Increased MUC4 staining has a positive correlation with higher tumor grades [85,130,131], but it is unclear if this can be associated with patient prognosis [85]. MUC16 stained positive in invasive micropapillary carcinoma of the breast in one study, but its prognostic correlation was not examined [132].

In prostate cancer, there is no consensus supporting MUC1 expression in normal and tumorigenic tissue, or an association with higher Gleason grades [50,85,111,133,134]. The lack of consensus can be attributed to the variability of alternate O-linked glyco-sylation and antibody specificity. However, in assessment of prostate cancer tissue using antibodies that can detect forms of MUC1 independent of glycosylation patterns (CT1 and 214D4 [135,136]), only 41% of normal prostate tissue samples were positive for MUC1, while it was only present in 26% of prostate tumor samples [137]. For MUC4, one study showed expression levels were downregulated in prostate cancer tissue, with 84.2% of tissue from normal cases stained positive for MUC4 compared with only 26.3% of the cancer cases [51].

MUC16 cancer-associated glycoforms (Tn and STn) and elevated serum levels are detected in more than 80% of epithelial ovarian cancer patients, making it useful for early disease detection, diagnostic staging and determining therapeutic treatment success [138142]. However, aberrant MUC16 expression has not been correlated with patient prognosis [86].

Although both MUC1 and MUC4 are expressed in normal and cancerous lung tissue [143], MUC4 expression is reduced in non-small-cell carcinomas (NSCLC), whereas MUC1 expression remains constant [144]. Detection of MUC1 with glycosylation-independent antibodies [145147] and depolarized localization are both considered poor prognostic markers [93]. Interestingly, one study found detection of MUC1 using a TF or Tn antigen-spe-cific antibody correlated with a more favorable prognosis [148]. Similarly, a possible association between elevated levels of MUC4 and longer survival rates for stages I and II NSCLC adeno-carcinoma patients has been suggested [149]. Conversely, in small-sized lung adenocarcinoma patients, high MUC4 expression has a lower survival rate [94,144].

Transmembrane mucins as therapeutic targets

Given their abundance and overexpression in many adeno-carcinomas, and the post-translational alteration in their glyco-sylation patterns, it is not surprising that transmembrane mucins have been investigated as potential targets for therapies. While there are currently no mucin-targeted therapies in clinical use, several MUC1 therapies are in clinical trials. Potential applications using nanotechnology-based therapies targeting mucins are also being examined in this regard (see Figure 2).

An external file that holds a picture, illustration, etc.
Object name is nihms340062f2.jpg
Transmembrane mucins as therapeutic targets

(A) Antibody-based therapies include radioisotopes (yellow star shapes) and drugs (blue ovals) conjugated to antibodies (green Y-shapes) recognizing the ectodomains of mucins. (B) Peptides and small-molecule drugs (turquoise oval) can block the active cytoplasmic domain of MUC1, thus preventing its localization to the mitochondria or nucleus. (C) Immunotherapies designed to stimulate the body’s immune response and induce T cells (blue amoeboid shape) that target mucins. (D) Targeted nanotherapies use antibodies or aptamers (blue hook shapes) that recognize the ectodomains of mucins to deliver gold nanoparticles (large orange circles) or quantum dots. For details, see text.

Antibody targeting

Antibodies targeting receptors that have prominent roles in cancer have been developed and US FDA approved for clinical use (e.g., Her2 [Herceptin®] and EGFR [cetuximab]). A number of antibodies have been raised against MUC1, and over 200 references in the literature describe anti-MUC1 antibodies. The VNTR region is extremely immunogenic, especially the APDTR region [150], and therefore leads to the development of a series of monoclonal antibodies targeting the N-terminus that are reactive with various epitopes of the domain [151].

While promising in animal models and preclinical studies, no MUC1 antibody therapy has successfully completed all phases of clinical trials. One humanized antibody, HMFG1, which recognizes the PDTR motif within the VNTR region, has been formulated in various treatments (in combination with cyto-toxic drugs) by the company Antisoma plc for the treatment of breast cancer [152]. Radiotherapies conjugated to HMFG1 [153], among other antibodies [154], have had disappointing outcomes, with no improved survival of the treated patients compared with the untreated patients. A Phase III clinical trial with Y90-labeled HMFG1 revealed that leakage of the antibody from the peritoneum and limited radiation treatment within the single dose were potential reasons for the failure of their trial for the treatment of ovarian cancer [155]. Promising early clinical studies have been reported recently by Immunomedics, Inc. for a combination of 90Y-PAM4 antibody to MUC1 (clivatuzumab) treatment with gemcitabine for advanced pancreatic cancer [156], although previous studies using this antibody were unsuccessful [157]. In addition, PAM4 has been shown to be effective in a blood assay for diagnosing early-stage pancreatic cancer [158].

Mucin antibodies have also been investigated for targeted drug delivery. Two MUC16 antibodies, one recognizing a non-repeat epitope in the extracellular domain and one recognizing the VNTR region, were conjugated to cytotoxic drugs. The antibody–drug conjugate targeted to the repeat region was more effi-cacious in a tumor mouse model than its non-repeat-targeting counterpart [159]. An antibody specific to the α/β junction of MUC1, the site of autocatalytic cleavage, was conjugated to an exotoxin that was only effective when it is internalized. In vivo results in a mouse model with human breast cancer cell lines show tumor suppression, indicating the antibody–toxin conjugates were intracellularized for the exotoxin to take effect [160].

Most of the antibody-targeted therapies have focused on the N-terminus of mucins, primarily because they are physically accessible to the therapy, and for their ability to target the altered glycoforms with an array of antibodies to choose from. However, these therapies must overcome the large pool of circulating shed N-terminal subunits for antibodies to reach the surface of carcinoma cells [83]. Furthermore, tumors are heterogenous tissues, as are the populations of MUC1 glycoforms present in these cancerous cells. Targeting more than one membrane-bound mucin and/or multiple epitopes of the same mucin at one time may prove to be a more effective therapeutic approach. In addition, antibodies recognizing the carboxy-terminal of mucins, which are unaffected by specific glycoform epitopes, may also hold promise for diagnostic and therapeutic applications [161].

Drug targets

The cytoplasmic domain (CD) of MUC1 plays an important role in cell signaling during cancer progression. MUC1-CD binds β-catenin, while also inhibiting the degradation of the EGFR. A peptide, MUC1 inhibitory peptide, designed to block the intracellular interactions between MUC1/β-catenin and MUC1/EGFR by acting as a binding decoy was used in vivo in mice. Mouse models treated with peptide, MUC1 inhibitory peptide displayed tumor regression and significant inhibition of tumor growth rate [162].

In cancer cells where MUC1 is overexpressed, MUC1-CD accumulates in the cytosol where it can localize to the mitochondria and nucleus [163]. Another peptide, GO-201, was designed to block the region where MUC1-CD is believed to dimerize/oligomerize and thus prevent its translocation. Treatment with GO-201 resulted in growth arrest and death of human breast cancer cells in a xenograft mouse model [164]. Furthermore, this peptide has shown high efficacy in a xenograft prostate cancer mouse model based on the MUC1-expressing prostate cancer cells, demonstrating complete tumor regression and prolonged lack of recurrence [165]. When non-MUC1-expressing prostate cancer cells were treated with GO-201, there was no evidence of cell death in vitro, as was the case for MUC1-positive cells, underlining the targeting specificity of the peptide [165]. This therapy is currently in Phase I trials in patients with advanced solid tumors [201].

Interestingly, small molecules found in nature appear to be effective in blocking MUC1-CD dimerization. Apigenin, a flavanoid found in leafy plants and vegetables, has been shown to interfere with the dimerization of the C-terminus of MUC1, thus blocking its localization to the nucleus. Apigenin-induced suppression of MUC1 C-terminal expression increased apoptosis and reduced clonogenic survival [166]. Similarly, the small molecule thymoqui-none, found in the seeds of Nigella sativa, reduces MUC4 expression in vitro. The decrease in MUC4 expression correlated with an increase in apoptosis, decreased motility and decreased migration in pancreatic cancer cells [167]. These small molecules are part of a larger class of natural products, which have been investigated for their anticancer properties.

Vaccines & immunotherapy

Of all the MUC1-targeted therapies in clinical trials, only two are in Phase III trials. Both are vaccines. The L-BLP25 liposome vaccine is designed to stimulate the body’s immune system and induce T cells that target cells expressing MUC1. The vaccine consists of a 25-amino-acid peptide containing the VNTR region and a palmi-toyl lysine residue at the carboxy terminal to enhance the incorporation of the lipopeptide into the liposome particle. Currently, it is in Phase III trials for NSCLC, which represents 80% of all lung cancers. A Phase IIb trial for stage IIIb/IV demonstrated that NSCLC patients who received the vaccine had a median survival of 30.6 months, versus 13.3 months [168]. L-BLP25 also shows promise for prostate cancer. The mean prostate-specific antigen doubling time in patients increased from 8.9 months at the onset of the trial to 12.2 months at the end of the study [169]. In addition, L-BLP25 is in Phase III trials for hormone-sensitive breast cancer [1].

The vaccine TG4010 (modified vaccinia virus Ankara-MUC1-IL2), a modified vaccinia virus Ankara strain based on a recombinant vaccinia virus expressing MUC1 antigen and the human cytokine IL-2, was also tested on patients with NSCLC [170]. During a Phase IIB trial, the 6-month survival rate increased to 17.1 months versus 11.3 months for patients who had normal levels of activated natural killer cells.

Another virus-based vaccine, PANVAC, which targeted carcino-embryonic antigen and MUC1, showed promise early on in clinical trials. In Phase I, patients with advanced pancreatic cancer who generated vaccine-specific T-cell responses had a median survival of 15.1 months compared with those without T-cell reactivity (3.9 months) [171]. A pilot study of PANVAC also showed promise for ovarian and breast cancer [172]. However, a Phase III randomized, controlled clinical trial of PANVAC for the treatment of patients with advanced pancreatic cancer did not meet its primary efficacy end point of improving overall survival compared with palliative chemotherapy or best supportive care [202]. PANVAC is one of many MUC1 immunotherapies to fail in a human clinical trial. Unfortunately, most of these attempts fail to translate from preclinical success into clinical efficacy [173].

Nanotherapies

Biological events, including those involved in cancer, occur on the nanoscale. If these processes can be detected or targeted as they happen, cancer could be diagnosed and treated at earlier stages. In addition, with this molecular precision, tumors can be targeted and destroyed with minimal harm to surrounding cells. As of yet, there are few studies using nanoscale entities that target membrane-bound mucins for diagnosis or treatment. One study [174] combined an imaging agent, quantum dots (QDs), conjugated to a DNA aptamer that was specific to the MUC1 glycoforms found on cancer cells [175]. This QD conjugate was also bound to doxorubicin via a pH-sensitive hydrazone bond, allowing the drug to be released after being intracellularized. In vivo mouse studies showed the QD-MUC1 aptamer accumulated selectively in tumor cells versus QD alone, indicating an active tumor-targeting mechanism in addition to the enhanced permeation and retention effect that allows most nanoparticles to passively target tumors through leaky vasculature. The release of doxorubicin, which quenches the fluorescence of QDs, was monitored in vitro via fluorescence under acidic conditions. This could provide a useful in vivo monitoring tool to measure doxorubicin release over time.

Another study used MUC1 to target pancreatic cancer cell xenografts in mouse models using gold nanoparticles conjugated to an anti-MUC1, PAM4. Radiofrequency radiation was used to trigger a hyperthermic effect from the gold particles, causing the cancer cells, which had a selectively higher uptake of gold nanoparticles than healthy cells, to be destroyed [176].

Recently, MUC1-expressing cells have been selectively killed in vitro by targeted plasmonic nanobubbles (PNBs) [177]. PNBs are transient lethal events generated by the laser irradiation of clusters of gold nanoparticles. The mechanical impact of these bubbles disrupts the cellular membrane and results in cell death. MUC1-targeted gold nanoparticles specifically bind to MUC1-expressing cells over cells that do not express MUC1. PNBs generated from anti-MUC1-gold particles were large enough to be cytotoxic. By comparison, PNBs targeting EGFR were also studied and resulted in smaller, less damaging bubbles, indicating that MUC1 was a superior targeting agent.

Expert commentary

Membrane-tethered mucins are extremely large cell surface components that play protective roles at the apical surfaces of normal epithelia and include MUC1, MUC4 and MUC16. Much is known about the normal patterns of distribution of these glycoproteins. Membrane-tethered mucins are often over-expressed in many cancers, but the heavy reliance on the use of glycoform-specific mucin antibodies in many studies may confuse interpretation. With regard to MUC1, much has been learned about gene regulation expression, including control by various cytokine-stimulated transcription factors and steroid hormone receptor family members. Information on MUC4 regulation is more limited and virtually nothing is known about MUC16 in this regard. More than one mucin can be expressed by a given cell in both normal and pathological contexts. Thus, it is important to understand if these genes are regulated coordinately and can be targeted as a group, or require individual therapeutic approaches.

Five-year view

There are potential medical benefits associated with being able to control membrane-tethered mucin expression. Increasing expression should improve mucosal barrier functions in individuals at risk of infections, or enhance other protective functions afforded by these glycoproteins. By contrast, reduction of mucin expression would be desirable in other instances, for example, in cancer therapies. Developing a more comprehensive understanding of the regulation of the major membrane-tethered mucin genes should reveal if they can be controlled as a group pharmacologically. Mucin-reducing therapies are likely to make tumors more susceptible to therapies with cytotoxic agents currently employed in cancer treatments. Few candidate agents to reduce expression of membrane-tethered mucins exist, although PPAR-γ agonists show some promise in this regard. Epitopes within the tandem repeat domains have been employed as agents to stimulate the host immune system to respond to mucin-expressing tumors; however, in spite of promising results in animal studies, human trials using this approach have, to date, been disappointing. More work is necessary to determine why the responses vary so much among species. Finally, available mucin-specific antibodies are currently being used to direct nanoparticles with cytotoxic payloads or cytolytic capabilities to selectively destroy mucin overexpressing tumors. These studies are at a primitive stage, but are certainly worth exploring. Systemic delivery of mucin-directed nanoparticles may be problematic for various reasons. Nonetheless, it should be possible in certain instances to deliver these agents directly to the tumor sites, for example, endome-trial carcinomas. The high accessibility of membrane-tethered mucins at the cell surface makes them excellent candidates for such targeting approaches.

Key issues

  • Because mucin expression is regulated by numerous factors and is dependent on cell and tissue type, it would be worthwhile to determine if the regulation of mucin expression is coordinated or distinct for each mucin gene.

  • Development of therapies that enhance or reduce mucin expression may prove to be beneficial.

  • The efficacy of peroxisome proliferator-activated receptor-γ agonists as general therapies in order to reduce mucin expression needs to be evaluated.

  • Combining mucin reduction therapies with conventional chemotherapies could potentially improve the efficacy of mucin-expressing cancer treatments.

  • Mucins show great promise as targets for immunotherapies and nanotherapies.

Acknowledgments

The authors thank Mary C Farach-Carson and members of the Carson and Farach-Carson laboratories for many helpful discussions. We are deeply appreciative of the excellent secretarial assistance of Sharron Kingston.

Footnotes

For reprint orders, please contact moc.sweiver-trepxe@stnirper

Financial & competing interests disclosure

The authors were supported by NIH grant R01HD029963 (to Daniel D Carson), NIH grant P50CA098258 NCI Uterine SPORE Pilot Project (to Daniel D Carson) and NIH grant U54 CA151668 Pilot Project (to Pamela Constantinou). The authors have no other relevant affiliations or financial in involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

1. Kufe DW. Mucins in cancer: function, prognosis and therapy. Nat Rev Cancer. 2009;9(12):874–885. [Europe PMC free article] [Abstract] [Google Scholar]
2. Hollingsworth MA, Swanson BJ. Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer. 2004;4(1):45–60. [Abstract] [Google Scholar]
3. Choudhury A, Moniaux N, Ringel J, et al. Alternate splicing at the 3′-end of the human pancreatic tumor-associated mucin MUC4 cDNA. Teratog Carcinog Mutagen. 2001;21(1):83–96. [Abstract] [Google Scholar]
4. Wreschner DH, Hareuveni M, Tsarfaty I, et al. Human epithelial tumor antigen cDNA sequences. Differential splicing may generate multiple protein forms. Eur J Biochem. 1990;189(3):463–473. [Abstract] [Google Scholar]
5. Ligtenberg MJ, Kruijshaar L, Buijs F, van Meijer M, Litvinov SV, Hilkens J. Cell-associated episialin is a complex containing two proteins derived from a common precursor. J Biol Chem. 1992;267(9):6171–6177. [Abstract] [Google Scholar]
6. Julian J, Carson DD. Formation of MUC1 metabolic complex is conserved in tumor-derived and normal epithelial cells. Biochem Biophys Res Commun. 2002;293(4):1183–1190. [Abstract] [Google Scholar]
7. Sheng Z, Wu K, Carraway KL, Fregien N. Molecular cloning of the transmembrane component of the 13762 mammary adenocarcinoma sialomucin complex. A new member of the epidermal growth factor superfamily. J Biol Chem. 1992;267(23):16341–16346. [Abstract] [Google Scholar]
8. Thathiah A, Blobel CP, Carson DD. Tumor necrosis factor-alpha converting enzyme/ADAM 17 mediates MUC1 shedding. J Biol Chem. 2003;278(5):3386–3394. [Abstract] [Google Scholar]
9. Thathiah A, Carson DD. MT1-MMP mediates MUC1 shedding independent of TACE/ADAM17. Biochem J. 2004;382(Pt 1):363–373. [Europe PMC free article] [Abstract] [Google Scholar]
10. Tian E, Ten Hagen KG. Recent insights into the biological roles of mucin-type O-glycosylation. Glycoconj J. 2009;26(3):325–334. [Europe PMC free article] [Abstract] [Google Scholar]
11. Ten Hagen KG, Fritz TA, Tabak LA. All in the family: the UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferases. Glycobiology. 2003;13(1):R1–R16. [Abstract] [Google Scholar]
12. Varki A. Selectins and other mammalian sialic acid-binding lectins. Curr Opin Cell Biol. 1992;4(2):257–266. [Abstract] [Google Scholar]
13. Sperandio M, Gleissner CA, Ley K. Glycosylation in immune cell trafficking. Immunol Rev. 2009;230(1):97–113. [Europe PMC free article] [Abstract] [Google Scholar]
14. Ramphal R, Arora SK. Recognition of mucin components by Pseudomonas aeruginosa. Glycoconj J. 2001;18(9):709–713. [Abstract] [Google Scholar]
15. Evans DJ, Jr, Evans DG. Helicobacter pylori adhesins: review and perspectives. Helicobacter. 2000;5(4):183–195. [Abstract] [Google Scholar]
16. Hattrup CL, Gendler SJ. Structure and function of the cell surface (tethered) mucins. Annu Rev Physiol. 2008;70:431–457. [Abstract] [Google Scholar]
17. Gendler SJ, Lancaster CA, Taylor-Papadimitriou J, et al. Molecular cloning and expression of human tumor-associated polymorphic epithelial mucin. J Biol Chem. 1990;265(25):15286–15293. [Abstract] [Google Scholar]
18. Hanisch FG, Muller S. MUC1: the polymorphic appearance of a human mucin. Glycobiology. 2000;10(5):439–449. [Abstract] [Google Scholar]
19. Kui Wong N, Easton RL, Panico M, et al. Characterization of the oligosaccharides associated with the human ovarian tumor marker CA125. J Biol Chem. 2003;278(31):28619–28634. [Abstract] [Google Scholar]
20. Wesseling J, van der Valk SW, Hilkens J. A mechanism for inhibition of E-cadherin-mediated cell–cell adhesion by the membrane-associated mucin episialin/MUC1. Mol Biol Cell. 1996;7(4):565–577. [Europe PMC free article] [Abstract] [Google Scholar]
21. Komatsu M, Carraway CA, Fregien NL, Carraway KL. Reversible disruption of cell–matrix and cell–cell interactions by overexpression of sialomucin complex. J Biol Chem. 1997;272(52):33245–33254. [Abstract] [Google Scholar]
22. Carson DD, Julian J, Lessey BA, Prakobphol A, Fisher SJ. MUC1 is a scaffold for selectin ligands in the human uterus. Front Biosci. 2006;11:2903–2908. [Abstract] [Google Scholar]
23. Gubbels JA, Felder M, Horibata S, et al. MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells. Mol Cancer. 2010;9:11. [Europe PMC free article] [Abstract] [Google Scholar]
24. Belisle JA, Horibata S, Jennifer GA, et al. Identification of Siglec-9 as the receptor for MUC16 on human NK cells, B cells, and monocytes. Mol Cancer. 2010;9:118. [Europe PMC free article] [Abstract] [Google Scholar]
25. Rump A, Morikawa Y, Tanaka M, et al. Binding of ovarian cancer antigen CA125/MUC16 to mesothelin mediates cell adhesion. J Biol Chem. 2004;279(10):9190–9198. [Abstract] [Google Scholar]
26. Carraway CA, Carraway KL. Sequestration and segregation of receptor kinases in epithelial cells: implications for ErbB2 oncogenesis. Sci STKE. 2007;(381):re3. [Abstract] [Google Scholar]
27. Carson DD. The cytoplasmic tail of MUC1: a very busy place. Sci Signal. 2008;1(27):pe35. [Abstract] [Google Scholar]
28. Vasir B, Avigan D, Wu, et al. Dendritic cells induce MUC1 expression and polarization on human T cells by an IL-7-dependent mechanism. J Immunol. 2005;174:2376–2386. [Abstract] [Google Scholar]
29. Vandewielvankemenade E, Ligtenberg MJL, Deboer AJ, et al. Episialin (Muc1) inhibits cytotoxic lymphocyte-target cell-interaction. J Immunol. 1993;151(2):767–776. [Abstract] [Google Scholar]
30. Komatsu M, Yee L, Carraway KL. Overexpression of sialomucin complex, a rat homologue of MUC4, inhibits tumor killing by lymphokine-activated killer cells. Cancer Res. 1999;59(9):2229–2236. [Abstract] [Google Scholar]
31. Agrawal B, Krantz MJ, Reddish MA, Longenecker BM. Cancer-associated MUC1 mucin inhibits human T-cell proliferation, which is reversible by IL-2. Nat Med. 1998;4(1):43–49. [Abstract] [Google Scholar]
32. Patankar MS, Jing Y, Morrison JC, et al. Potent suppression of natural killer cell response mediated by the ovarian tumor marker CA125. Gynecol Oncol. 2005;99(3):704–713. [Abstract] [Google Scholar]
33. Spicer AP, Rowse GJ, Lidner TK, Gendler SJ. Delayed mammary tumor progression in Muc-1 null mice. J Biol Chem. 1995;270(50):30093–30101. [Abstract] [Google Scholar]
34. Kardon R, Price RE, Julian J, et al. Bacterial conjunctivitis in Muc1 null mice. Invest Ophthalmol Vis Sci. 1999;40(7):1328–1335. [Abstract] [Google Scholar]
35. Danjo Y, Hazlett LD, Gipson IK. C57BL/6 mice lacking Muc1 show no ocular surface phenotype. Invest Ophthalmol Vis Sci. 2000;41(13):4080–4084. [Abstract] [Google Scholar]
36. Cheon DJ, Wang Y, Deng JM, et al. CA125/MUC16 is dispensable for mouse development and reproduction. PLoS One. 2009;4(3):e4675. [Europe PMC free article] [Abstract] [Google Scholar]
37. Jonckheere N, Van Seuningen I. The membrane-bound mucins: from cell signalling to transcriptional regulation and expression in epithelial cancers. Biochimie. 2010;92(1):1–11. [Abstract] [Google Scholar]
38. Gendler SJ. MUC1, the renaissance molecule. J Mammary Gland Biol Neoplasia. 2001;6(3):339–353. [Abstract] [Google Scholar]
39. Brayman M, Thathiah A, Carson DD. MUC1: a multifunctional cell surface component of reproductive tissue epithelia. Reprod Biol Endocrinol. 2004;2:4. [Europe PMC free article] [Abstract] [Google Scholar]
40. Dent GA, Civalier CJ, Brecher ME, Bentley SA. MUC1 expression in hematopoietic tissues. Am J Clin Pathol. 1999;111(6):741–747. [Abstract] [Google Scholar]
41. Chang JF, Zhao HL, Phillips J, Greenburg G. The epithelial mucin, MUC1, is expressed on resting T lymphocytes and can function as a negative regulator of T cell activation. Cell Immunol. 2000;201(2):83–88. [Abstract] [Google Scholar]
42. Agrawal B, Krantz MJ, Parker J, Longenecker BM. Expression of MUC1 mucin on activated human T cells: implications for a role of MUC1 in normal immune regulation. Cancer Res. 1998;58(18):4079–4081. [Abstract] [Google Scholar]
43. Franke FE, Kraus S, Eiermann C, Pauls K, Lalani EN, Bergmann M. MUC1 in normal and impaired spermatogenesis. Mol Hum Reprod. 2001;7(6):505–512. [Abstract] [Google Scholar]
44. Chaturvedi P, Singh AP, Batra SK. Structure, evolution, and biology of the MUC4 mucin. FASEB J. 2008;22(4):966–981. [Europe PMC free article] [Abstract] [Google Scholar]
45. Argueso P, Spurr-Michaud S, Russo CL, Tisdale A, Gipson IK. MUC16 mucin is expressed by the human ocular surface epithelia and carries the H185 carbohydrate epitope. Invest Ophthalmol Vis Sci. 2003;44(6):2487–2495. [Abstract] [Google Scholar]
46. Davies JR, Kirkham S, Svitacheva N, Thornton DJ, Carlstedt I. MUC16 is produced in tracheal surface epithelium and submucosal glands and is present in secretions from normal human airway and cultured bronchial epithelial cells. Int J Biochem Cell Biol. 2007;39(10):1943–1954. [Abstract] [Google Scholar]
47. Zeimet AG, Offner FA, Muller-Holzner E, et al. Peritoneum and tissues of the female reproductive tract as physiological sources of CA-125. Tumour Biol. 1998;19(4):275–282. [Abstract] [Google Scholar]
48. Zeimet AG, Muller-Holzner E, Marth C, Daxenbichler G, Dapunt O. Tumor marker CA-125 in tissues of the female reproductive tract and in serum during the normal menstrual cycle. Fertil Steril. 1993;59(5):1028–1035. [Abstract] [Google Scholar]
49. Nustad K, Lebedin Y, Lloyd KO, et al. Epitopes on CA 125 from cervical mucus and ascites fluid and characterization of six new antibodies. Third report from the ISOBM TD-1 workshop. Tumour Biol. 2002;23(5):303–314. [Abstract] [Google Scholar]
50. O’Connor JC, Julian J, Lim SD, Carson DD. MUC1 expression in human prostate cancer cell lines and primary tumors. Prostate Cancer Prostatic Dis. 2005;8(1):36–44. [Abstract] [Google Scholar]
51. Singh AP, Chauhan SC, Bafna S, et al. Aberrant expression of transmembrane mucins, MUC1 and MUC4, in human prostate carcinomas. Prostate. 2006;66(4):421–429. [Abstract] [Google Scholar]
52. Bast RC, Jr, Klug TL, St John E, et al. A radioimmunoassay using a monoclonal antibody to monitor the course of epithelial ovarian cancer. N Engl J Med. 1983;309(15):883–887. [Abstract] [Google Scholar]
53. Ginath S, Menczer J, Fintsi Y, Ben-Shem E, Glezerman M, Avinoach I. Tissue and serum CA125 expression in endometrial cancer. Int J Gynecol Cancer. 2002;12(4):372–375. [Abstract] [Google Scholar]
54. Carson DD, Dharmaraj N, Wang P. Transcriptional control of the expression of MUC1. Expert Rev Endocrinol Metab. 2008;3(4):463–471. [Abstract] [Google Scholar]
55. McNeer RR, Carraway CA, Fregien NL, Carraway KL. Characterization of the expression and steroid hormone control of sialomucin complex in the rat uterus: implications for uterine receptivity. J Cell Physiol. 1998;176(1):110–119. [Abstract] [Google Scholar]
56. Idris N, Carraway KL. Sialomucin complex (Muc4) expression in the rat female reproductive tract. Biol Reprod. 1999;61(6):1431–1438. [Abstract] [Google Scholar]
57. Lange C, Fernandez J, Shim D, Spurr-Michaud S, Tisdale A, Gipson IK. Mucin gene expression is not regulated by estrogen and/or progesterone in the ocular surface epithelia of mice. Exp Eye Res. 2003;77(1):59–68. [Abstract] [Google Scholar]
58. Idris N, Carraway KL. Regulation of sialomucin complex/Muc4 expression in rat uterine luminal epithelial cells by transforming growth factor-β: implications for blastocyst implantation. J Cell Physiol. 2000;185(2):310–316. [Abstract] [Google Scholar]
59. Gipson IK, Ho SB, Spurr-Michaud SJ, et al. Mucin genes expressed by human female reproductive tract epithelia. Biol Reprod. 1997;56(4):999–1011. [Abstract] [Google Scholar]
60. Gipson IK, Blalock T, Tisdale A, et al. MUC16 is lost from the uterodome (pinopode) surface of the receptive human endometrium: in vitro evidence that MUC16 is a barrier to trophoblast adherence. Biol Reprod. 2008;78(1):134–142. [Abstract] [Google Scholar]
61. Brayman MJ, Julian J, Mulac-Jericevic B, Conneely OM, Edwards DP, Carson DD. Progesterone receptor isoforms A and B differentially regulate MUC1 expression in uterine epithelial cells. Mol Endocrinol. 2006;20(10):2278–2291. [Abstract] [Google Scholar]
62. Wang P, Dharmaraj N, Brayman MJ, Carson DD. Peroxisome proliferator-activated receptor γ activation inhibits progesterone-stimulated human MUC1 expression. Mol Endocrinol. 2010;24(7):1368–1379. [Europe PMC free article] [Abstract] [Google Scholar]
63. Dharmaraj N, Wang P, Carson DD. Cytokine and progesterone receptor interplay in the regulation of MUC1 gene expression. Mol Endocrinol. 2010;24(12):2253–2266. [Europe PMC free article] [Abstract] [Google Scholar]
64. Perrais M, Pigny P, Ducourouble MP, et al. Characterization of human mucin gene MUC4 promoter: importance of growth factors and proinflammatory cytokines for its regulation in pancreatic cancer cells. J Biol Chem. 2001;276(33):30923–30933. [Abstract] [Google Scholar]
65. Andrianifahanana M, Singh AP, Nemos C, et al. IFN-γ-induced expression of MUC4 in pancreatic cancer cells is mediated by STAT-1 upregulation: a novel mechanism for IFN-γ response. Oncogene. 2007;26(51):7251–7261. [Abstract] [Google Scholar]
66. Jonckheere N, Perrais M, Mariette C, et al. A role for human MUC4 mucin gene, the ErbB2 ligand, as a target of TGF-β in pancreatic carcinogenesis. Oncogene. 2004;23(34):5729–5738. [Abstract] [Google Scholar]
67. Choudhury A, Singh RK, Moniaux N, El-Metwally TH, Aubert JP, Batra SK. Retinoic acid-dependent transforming growth factor-β 2-mediated induction of MUC4 mucin expression in human pancreatic tumor cells follows retinoic acid receptor-α signaling pathway. J Biol Chem. 2000;275(43):33929–33936. [Abstract] [Google Scholar]
68. Andrianifahanana M, Agrawal A, Singh AP, et al. Synergistic induction of the MUC4 mucin gene by interferon-γ and retinoic acid in human pancreatic tumour cells involves a reprogramming of signalling pathways. Oncogene. 2005;24(40):6143–6154. [Abstract] [Google Scholar]
69. Jonckheere N, Vincent A, Perrais M, et al. The human mucin MUC4 is transcriptionally regulated by caudal-related homeobox, hepatocyte nuclear factors, forkhead box A, and GATA endodermal transcription factors in epithelial cancer cells. J Biol Chem. 2007;282(31):22638–22650. [Abstract] [Google Scholar]
70. Piessen G, Jonckheere N, Vincent A, et al. Regulation of the human mucin MUC4 by taurodeoxycholic and taurochenodeoxycholic bile acids in oesophageal cancer cells is mediated by hepatocyte nuclear factor 1α Biochem J. 2007;402(1):81–91. [Europe PMC free article] [Abstract] [Google Scholar]
71. Albertsmeyer AC, Kakkassery V, Spurr-Michaud S, Beeks O, Gipson IK. Effect of pro-inflammatory mediators on membrane-associated mucins expressed by human ocular surface epithelial cells. Exp Eye Res. 2010;90(3):444–451. [Europe PMC free article] [Abstract] [Google Scholar]
72. Thapi A, Binder D, Roseales N, Yan XJ, Lash AE, Spriggs DR. Transcription factors that regulate MUC16 expression in ovarian cancer. Presented at: AACR 102nd Annual Meeting; Orlando, FL, USA. 2–6 April 2011. [Google Scholar]
73. Van Seuningen I, Vincent A. Mucins: a new family of epigenetic biomarkers in epithelial cancers. Expert Opin Med Diagnostics. 2009;3(4):411–442. [Abstract] [Google Scholar]
74. Smorodinsky N, Weiss M, Hartmann ML, et al. Detection of a secreted MUC1/SEC protein by MUC1 isoform specific monoclonal antibodies. Biochem Biophys Res Commun. 1996;228(1):115–121. [Abstract] [Google Scholar]
75. Zrihan-Licht S, Vos HL, Baruch A, et al. Characterization and molecular cloning of a novel MUC1 protein, devoid of tandem repeats, expressed in human breast cancer tissue. Eur J Biochem. 1994;224(2):787–795. [Abstract] [Google Scholar]
76. Baruch A, Hartmann M, Zrihan-Licht S, et al. Preferential expression of novel MUC1 tumor antigen isoforms in human epithelial tumors and their tumor-potentiating function. Int J Cancer. 1997;71(5):741–749. [Abstract] [Google Scholar]
77. Oosterkamp HM, Scheiner L, Stefanova MC, Lloyd KO, Finstad CL. Comparison of MUC-1 mucin expression in epithelial and non-epithelial cancer cell lines and demonstration of a new short variant form (MUC-1/Z) Int J Cancer. 1997;72(1):87–94. [Abstract] [Google Scholar]
78. Levitin F, Baruch A, Weiss M, et al. A novel protein derived from the MUC1 gene by alternative splicing and frameshifting. J Biol Chem. 2005;280(11):10655–10663. [Abstract] [Google Scholar]
79. Choudhury A, Moniaux N, Winpenny JP, Hollingsworth MA, Aubert JP, Batra SK. Human MUC4 mucin cDNA and its variants in pancreatic carcinoma. J Biochem. 2000;128(2):233–243. [Abstract] [Google Scholar]
80. Moniaux N, Escande F, Batra SK, Porchet N, Laine A, Aubert JP. Alternative splicing generates a family of putative secreted and membrane-associated MUC4 mucins. Eur J Biochem. 2000;267(14):4536–4544. [Abstract] [Google Scholar]
81. Briggs S, Price MR, Tendler SJ. Immune recognition of linear epitopes in peptide fragments of epithelial mucins. Immunology. 1991;73(4):505–507. [Abstract] [Google Scholar]
82. Hayes DF, Sekine H, Ohno T, Abe M, Keefe K, Kufe DW. Use of a murine monoclonal antibody for detection of circulating plasma DF3 antigen levels in breast cancer patients. J Clin Invest. 1985;75(5):1671–1678. [Europe PMC free article] [Abstract] [Google Scholar]
83. Kufe DW. Targeting the human MUC1 oncoprotein: a tale of two proteins. Cancer Biol Ther. 2008;7(1):81–84. [Abstract] [Google Scholar]
84. Nacht M, Ferguson AT, Zhang W, et al. Combining serial analysis of gene expression and array technologies to identify genes differentially expressed in breast cancer. Cancer Research. 1999;59(21):5464–5470. [Abstract] [Google Scholar]
85. Rakha EA, Boyce RWG, Abd El-Rehim D, et al. Expression of mucins (MUC1, MUC2, MUC3, MUC4, MUC5AC and MUC6) and their prognostic significance in human breast cancer. Mod Pathol. 2005;18(10):1295–1304. [Abstract] [Google Scholar]
86. Chauhan SC, Singh AP, Ruiz F, et al. Aberrant expression of MUC4 in ovarian carcinoma: diagnostic significance alone and in combination with MUC1 and MUC16 (CA125) Mod Pathol. 2006;19(10):1386–1394. [Abstract] [Google Scholar]
87. Nakamori S, Ota DM, Cleary KR, Shirotani K, Irimura T. MUC1 mucin expression as a marker of progression and metastasis of human colorectal carcinoma. Gastroenterology. 1994;106(2):353–361. [Abstract] [Google Scholar]
88. Hiraga Y, Tanaka S, Haruma K, et al. Immunoreactive MUC1 expression at the deepest invasive portion correlates with prognosis of colorectal cancer. Oncology. 1998;55(4):307–319. [Abstract] [Google Scholar]
89. Luttges J, Feyerabend B, Buchelt T, Pacena M, Kloppel G. The mucin profile of noninvasive and invasive mucinous cystic neoplasms of the pancreas. Am J Surg Pathol. 2002;26(4):466–471. [Abstract] [Google Scholar]
90. Kashiwagi H, Kijima H, Dowaki S, et al. DF3 expression in human gallbladder carcinoma: significance for lymphatic invasion. Int J Oncol. 2000;16(3):455–459. [Abstract] [Google Scholar]
91. Nitta T, Sugihara K, Tsuyama S, Murata F. Immunohistochemical study of MUC1 mucin in premalignant oral lesions and oral squamous cell carcinoma: association with disease progression, mode of invasion, and lymph node metastasis. Cancer. 2000;88(2):245–254. [Abstract] [Google Scholar]
92. Miyazaki K, Kurishima K, Kagohashi K, et al. Serum KL-6 levels in lung cancer patients with or without interstitial lung disease. J Clin Lab Anal. 2010;24(5):295–299. [Abstract] [Google Scholar]
93. Nagai S, Takenaka K, Sonobe M, Ogawa E, Wada H, Tanaka F. A novel classification of MUC1 expression is correlated with tumor differentiation and postoperative prognosis in non-small cell lung cancer. J Thorac Oncol. 2006;1(1):46–51. [Abstract] [Google Scholar]
94. Tsutsumida H, Goto M, Kitajima S, et al. MUC4 expression correlates with poor prognosis in small-sized lung adenocarcinoma. Lung Cancer. 2007;55(2):195–203. [Abstract] [Google Scholar]
95. Satoh S, Hinoda Y, Hayashi T, Burdick MD, Imai K, Hollingsworth MA. Enhancement of metastatic properties of pancreatic cancer cells by MUC1 gene encoding an anti-adhesion molecule. Int J Cancer. 2000;88(4):507–518. [Abstract] [Google Scholar]
96. Raina D, Ahmad R, Kumar S, et al. MUC1 oncoprotein blocks nuclear targeting of c-Abl in the apoptotic response to DNA damage. EMBO J. 2006;25(16):3774–3783. [Europe PMC free article] [Abstract] [Google Scholar]
97. Zhang K, Baeckstrom D, Brevinge H, Hansson GC. Secreted MUC1 mucins lacking their cytoplasmic part and carrying sialyl-Lewis a and x epitopes from a tumor cell line and sera of colon carcinoma patients can inhibit HL-60 leukocyte adhesion to E-selectin-expressing endothelial cells. J Cell Biochem. 1996;60(4):538–549. [Abstract] [Google Scholar]
98. Nath D, Hartnell A, Happerfield L, et al. Macrophage-tumour cell interactions: identification of MUC1 on breast cancer cells as a potential counter-receptor for the macrophage-restricted receptor, sialoadhesin. Immunology. 1999;98(2):213–219. [Abstract] [Google Scholar]
99. Komatsu M, Tatum L, Altman NH, Carraway CAC, Carraway KL. Potentiation of metastasis by cell surface sialomucin complex (rat MUC4), a multifunctional anti-adhesive glycoprotein. Int J Cancer. 2000;87(4):480–486. [Abstract] [Google Scholar]
100. Kondo K, Kohno N, Yokoyama A, Hiwada K. Decreased MUC1 expression induces E-cadherin-mediated cell adhesion of breast cancer cell lines. Cancer Res. 1998;58(9):2014–2019. [Abstract] [Google Scholar]
101. Wesseling J, Vandervalk SW, Vos HL, Sonnenberg A, Hilkens J. Episialin (MUC1) overexpression inhibits integrin-mediated cell-adhesion to extracellular-matrix components. J Cell Biol. 1995;129(1):255–265. [Europe PMC free article] [Abstract] [Google Scholar]
102. Carraway KL, Theodoropoulos G, Kozloski GA, Carraway CAC. Muc4/MUC4 functions and regulation in cancer. Future Oncol. 2009;5(10):1631–1640. [Europe PMC free article] [Abstract] [Google Scholar]
103. Workman HC, Sweeney C, Carraway KL. The membrane mucin Muc4 inhibits apoptosis induced by multiple insults via ErbB2-dependent and ErbB2-independent mechanisms. Cancer Res. 2009;69(7):2845–2852. [Europe PMC free article] [Abstract] [Google Scholar]
104. Comamala M, Pinard M, Theriault C, et al. Downregulation of cell surface CA125/MUC16 induces epithelial-to-mesenchymal transition and restores EGFR signalling in NIH:OVCAR3 ovarian carcinoma cells. Br J Cancer. 2011;104(6):989–999. [Europe PMC free article] [Abstract] [Google Scholar]
105. Gubbels JAA, Belisle J, Onda M, et al. Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol Cancer. 2006;5(1):50. [Europe PMC free article] [Abstract] [Google Scholar]
106. Seelenmeyer C, Wegehingel S, Lechner J, Nickel W. The cancer antigen CA125 represents a novel counter receptor for galectin-1. J Cell Sci. 2003;116(7):1305–1318. [Abstract] [Google Scholar]
107. Corfield AP, Myerscough N, Longman R, Sylvester P, Arul S, Pignatelli M. Mucins and mucosal protection in the gastrointestinal tract: new prospects for mucins in the pathology of gastrointestinal disease. Gut. 2000;47(4):589–594. [Europe PMC free article] [Abstract] [Google Scholar]
108. Parry S, Hanisch FG, Leir SH, et al. N-Glycosylation of the MUC1 mucin in epithelial cells and secretions. Glycobiology. 2006;16(7):623–634. [Abstract] [Google Scholar]
109. Baldus SE, Engelmann K, Hanisch FG. MUC1 and the MUCs: A family of human mucins with impact in cancer biology. Crit Rev Clin Lab Sci. 2004;41(2):189–231. [Abstract] [Google Scholar]
110. Croce MV, Isla-Larrain M, Tur R, Rabassa ME, Segal-Eiras A. Antigenic differences between metastatic cells in bone marrow and primary tumours and the anti-MUC1 humoral immune response induced in breast cancer patients. Clin Exp Metastasis. 2004;21(2):139–147. [Abstract] [Google Scholar]
111. Burke PA, Gregg JP, Bakhtiar B, et al. Characterization of MUC1 glycoprotein on prostate cancer for selection of targeting molecules. Int J Oncol. 2006;29(1):49–55. [Abstract] [Google Scholar]
112. Demichelis SO, Alberdi CG, Servi WJ, Isla-Larrain MT, Segal-Eiras A, Croce MV. Comparative immunohistochemical study of MUC1 and carbohydrate antigens in breast benign disease and normal mammary gland. Appl Immunohistochem Mol Morphol. 2010;18(1):41–50. [Abstract] [Google Scholar]
113. Wang PH, Li YF, Juang CM, et al. Altered mRNA expression of sialyltransferase in squamous cell carcinomas of the cervix. Gynecol Oncol. 2001;83(1):121–127. [Abstract] [Google Scholar]
114. Wang PH, Lo WL, Hsu CC, et al. Different enzyme activities of sialyltransferases in gynecological cancer cell lines. Eur J Gynaecol Oncol. 2002;23(3):221–226. [Abstract] [Google Scholar]
115. Zhu YT, Srivatana U, Ullah A, Gagneja H, Berenson CS, Lance P. Suppression of a sialyltransferase by antisense DNA reduces invasiveness of human colon cancer cells in vitro. Biochim Biophys Acta. 2001;1536(2–3):148–160. [Abstract] [Google Scholar]
116. Sewell R, Backstrom M, Dalziel M, et al. The ST6GalNAc-I sialyltransferase localizes throughout the golgi and is responsible for the synthesis of the tumor-associated Sialyl-Tn O-glycan in human breast cancer. J Biol Chem. 2006;281(6):3586–3594. [Abstract] [Google Scholar]
117. Picco G, Julien S, Brockhausen I, et al. Over-expression of ST3Gal-I promotes mammary tumorigenesis. Glycobiology. 2010;20(10):1241–1250. [Europe PMC free article] [Abstract] [Google Scholar]
118. Jun L, Yuanshu W, Yanying X, et al. Altered mRNA expressions of sialyltransferases in human gastric cancer tissues. Med Oncol. 2010 10.1007/s12032-010-9771-1. (Epub ahead of print) [Abstract] [CrossRef] [Google Scholar]
119. Vazquez-Martin C, Cuevas E, Gil-Martin E, Fernandez-Briera A. Correlation analysis between tumor-associated antigen sialyl-Tn expression and ST6GalNAc I activity in human colon adenocarcinoma. Oncology. 2004;67(2):159–165. [Abstract] [Google Scholar]
120. Bouanene H, Sahrawi W, Mokni M, et al. Correlation between heterogeneous expression of sialyltransferases and MUC16 in ovarian tumor tissues. Onkologie. 2011;34(4):165–169. [Abstract] [Google Scholar]
121. Gendler SJ. MUC1, the renaissance molecule. J Mammary Gland Biol. 2001;6(3):339–353. [Abstract] [Google Scholar]
122. Lee G, Ge BX, Huang TK, Zheng G, Duan JT, Wang IHY. Positive identification of CA215 pan cancer biomarker from serum specimens of cancer patients. Cancer Biomark. 2010;6(2):111–117. [Abstract] [Google Scholar]
123. Lau SK, Weiss LM, Chu PG. Differential expression of MUC1, MUC2, and MUC5AC in carcinomas of various sites: an immunohistochemical study. Am J Clin Pathol. 2004;122(1):61–69. [Abstract] [Google Scholar]
124. Schroeder JA, Al Masri A, Adriance MC, et al. MUC1 overexpression results in mammary gland tumorigenesis and prolonged alveolar differentiation. Oncogene. 2004;23(34):5739–5747. [Abstract] [Google Scholar]
125. Kesisis G, Kontovinis LF, Gennatas K, Kortsaris AH. Biological markers in breast cancer prognosis and treatment. J Buon. 2010;15(3):447–454. [Abstract] [Google Scholar]
126. Hayes DF, Bast RC, Desch CE, et al. Tumor marker utility grading system: a framework to evaluate clinical utility of tumor markers. J Natl Cancer Inst. 1996;88(20):1456–1466. [Abstract] [Google Scholar]
127. Walsh MD, Luckie SM, Cummings MC, Antalis TM, McGuckin MA. Heterogeneity of MUC1 expression by human breast carcinoma cell lines in vivo and in vitro. Breast Cancer Res Treat. 1999;58(3):255–266. [Abstract] [Google Scholar]
128. Al-Azawi D, Kelly G, Myers E, et al. CA 15–13 is predictive of response and disease recurrence following treatment in locally advanced breast cancer. BMC Cancer. 2006;6:220. [Europe PMC free article] [Abstract] [Google Scholar]
129. Kumpulainen EJ, Keskikuru RJ, Johansson RT. Serum tumor marker CA 15.3 and stage are the two most powerful predictors of survival in primary breast cancer. Breast Cancer Res Treat. 2002;76(2):95–102. [Abstract] [Google Scholar]
130. Workman HC, Miller JK, Ingalla EQ, et al. The membrane mucin MUC4 is elevated in breast tumor lymph node metastases relative to matched primary tumors and confers aggressive properties to breast cancer cells. Breast Cancer Research. 2009;11(5):R70. [Europe PMC free article] [Abstract] [Google Scholar]
131. Mukhopadhyay P, Chakraborty S, Ponnusamy MP, Lakshmanan I, Jain M, Batra SK. Mucins in the pathogenesis of breast cancer: implications in diagnosis, prognosis and therapy. Biochim Biophys Acta. 2011;1815(2):224–240. [Europe PMC free article] [Abstract] [Google Scholar]
132. Moritani S, Ichihara S, Hasegawa M, et al. Serous papillary adenocarcinoma of the female genital organs and invasive micropapillary carcinoma of the breast. Are WT1, CA125, and GCDFP-15 useful in differential diagnosis? Hum Pathol. 2008;39(5):666–671. [Abstract] [Google Scholar]
133. Cozzi PJ, Wang J, Delprado W, et al. Muc1, Muc2, Muc4, Muc5ac and Muc6 expression in the progression of prostate cancer. Clin Exp Metastas. 2005;22(7):565–573. [Abstract] [Google Scholar]
134. Kirschenbaum A, Itzkowitz SH, Wang JP, Yao S, Eliashvili M, Levine AC. MUC1 expression in prostate carcinoma: correlation with grade and stage. Mol Urol. 1999;3(3):163–167. [Abstract] [Google Scholar]
135. Pemberton L, Taylor-Papadimitriou J, Gendler SJ. Antibodies to the cytoplasmic domain of the MUC1 mucin show conservation throughout mammals. Biochem Biophys Res Commun. 1992;185(1):167–175. [Abstract] [Google Scholar]
136. Price MR, Rye PD, Petrakou E, et al. Summary report on the ISOBM TD-4 Workshop: analysis of 56 monoclonal antibodies against the MUC1 mucin. San Diego, Calif., November 17–23, 1996. Tumour Biol. 1998;19(Suppl 1):1–20. [Abstract] [Google Scholar]
137. O’Connor JC, Julian J, Lim SD, Carson DD. MUC1 expression in human prostate cancer cell lines and primary tumors. Prostate Cancer Prostatic Dis. 2005;8(1):36–44. [Abstract] [Google Scholar]
138. Capstick V, Maclean GD, Suresh MR, et al. Clinical evaluation of a new two-site assay for CA125 antigen. Int J Biol Markers. 1991;6(2):129–135. [Abstract] [Google Scholar]
139. Bast RC, Xu FJ, Yu YH, Barnhill S, Zhang Z, Mills GB. Ca 125: the past and the future. Int J Biol Marker. 1998;13(4):179–187. [Abstract] [Google Scholar]
140. Bafna S, Kaur S, Batra SK. Membrane-bound mucins: the mechanistic basis for alterations in the growth and survival of cancer cells. Oncogene. 2010;29(20):2893–2904. [Europe PMC free article] [Abstract] [Google Scholar]
141. Van Elssen CH, Frings PW, Bot FJ, et al. Expression of aberrantly glycosylated Mucin-1 in ovarian cancer. Histopathology. 2010;57(4):597–606. [Abstract] [Google Scholar]
142. Budiu RA, Mantia-Smaldone G, Elishaev E, et al. Soluble MUC1 and serum MUC1-specific antibodies are potential prognostic biomarkers for platinum-resistant ovarian cancer. Cancer Immunol Immunother. 2011;60(7):975–984. [Abstract] [Google Scholar]
143. Seregni E, Botti C, Lombardo C, et al. Pattern of mucin gene expression in normal and neoplastic lung tissues. Anticancer Res. 1996;16(4B):2209–2213. [Abstract] [Google Scholar]
144. Lopez-Ferrer A, Curull V, Barranco C, et al. Mucins as differentiation markers in bronchial epithelium. Squamous cell carcinoma and adenocarcinoma display similar expression patterns. Am J Respir Cell Mol Biol. 2001;24(1):22–29. [Abstract] [Google Scholar]
145. Guddo F, Giatromanolaki A, Koukourakis MI, et al. MUC1 (episialin) expression in non-small cell lung cancer is independent of EGFR and c-erbB-2 expression and correlates with poor survival in node positive patients. J Clin Pathol. 1998;51(9):667–671. [Europe PMC free article] [Abstract] [Google Scholar]
146. Ohgami A, Tsuda T, Osaki T, et al. MUC1 mucin mRNA expression in stage I lung adenocarcinoma and its association with early recurrence. Ann Thorac Surg. 1999;67(3):810–814. [Abstract] [Google Scholar]
147. MacDermed DM, Khodarev NN, Pitroda SP, et al. MUC1-associated proliferation signature predicts outcomes in lung adenocarcinoma patients. BMC Med Genomics. 2010;3:16. [Europe PMC free article] [Abstract] [Google Scholar]
148. Kuemmel A, Single K, Bittinger F, et al. TA-MUC1 epitope in non-small cell lung cancer. Lung Cancer. 2009;63(1):98–105. [Abstract] [Google Scholar]
149. Kwon KY, Ro JY, Singhal N, et al. MUC4 expression in non-small cell lung carcinomas: relationship to tumor histology and patient survival. Arch Pathol Lab Med. 2007;131(4):593–598. [Abstract] [Google Scholar]
150. Mollick JA, Hodi FS, Soiffer RJ, Nadler LM, Dranoff G. MUC1-like tandem repeat proteins are broadly immunogenic in cancer patients. Cancer Immun. 2003;3:3. [Abstract] [Google Scholar]
151. Singh R, Bandyopadhyay D. MUC1: a target molecule for cancer therapy. Cancer Biol Ther. 2007;6(4):481–486. [Abstract] [Google Scholar]
152. Beatson RE, Taylor-Papadimitriou J, Burchell JM. MUC1 immunotherapy. Immunotherapy. 2010;2(3):305–327. [Abstract] [Google Scholar]
153. Verheijen RH, Massuger LF, Benigno BB, et al. Phase III trial of intraperitoneal therapy with yttrium-90-labeled HMFG1 murine monoclonal antibody in patients with epithelial ovarian cancer after a surgically defined complete remission. J Clin Oncol. 2006;24(4):571–578. [Abstract] [Google Scholar]
154. Buckman R, De Angelis C, Shaw P, et al. Intraperitoneal therapy of malignant ascites associated with carcinoma of ovary and breast using radioiodinated monoclonal antibody 2G3. Gynecol Oncol. 1992;47(1):102–109. [Abstract] [Google Scholar]
155. Oei AL, Massuger LF, Oyen WJ. Extraperitoneal leakage as a possible explanation for failure of one-time intraperitoneal treatment in ovarian cancer. Cancer Biother Radiopharm. 2007;22(4):508–514. [Abstract] [Google Scholar]
156. Ocean AJ, Guarino MJ, Pennington KL, et al. Activity of fractionated radioimmunotherapy with clivatuzumab tetraxetan combined with low-dose gemcitabine (Gem) in advanced pancreatic cancer (APC) J Clin Oncol. 2011;29(4 Suppl):240. [Google Scholar]
157. Gulec SA, Cohen SJ, Pennignton KL, et al. Treatment of advanced pancreatic carcinoma with 90Y-clivatuzumab tetraxetan: a Phase I single-dose escalation trial. Clin Cancer Res. 2011;17(12):4091–4100. [Europe PMC free article] [Abstract] [Google Scholar]
158. Gold DV, Goggins M, Modrak DE, et al. Detection of early-stage pancreatic adenocarcinoma. Cancer Epidemiol Biomarkers Prev. 2010;19(11):2786–2794. [Europe PMC free article] [Abstract] [Google Scholar]
159. Chen Y, Clark S, Wong T, et al. Armed antibodies targeting the mucin repeats of the ovarian cancer antigen, MUC16, are highly efficacious in animal tumor models. Cancer Res. 2007;67(10):4924–4932. [Abstract] [Google Scholar]
160. Rubinstein DB, Karmely M, Pichinuk E, et al. The MUC1 oncoprotein as a functional target: immunotoxin binding to α/β junction mediates cell killing. Int J Cancer. 2009;124(1):46–54. [Abstract] [Google Scholar]
161. Dharma Rao T, Park KJ, Smith-Jones P, et al. Novel monoclonal antibodies against the proximal (carboxy-terminal) portions of MUC16. Appl Immunohistochem Mol Morphol. 2010;18(5):462–472. [Europe PMC free article] [Abstract] [Google Scholar]
162. Bitler BG, Menzl I, Huerta CL, et al. Intracellular MUC1 peptides inhibit cancer progression. Clin Cancer Res. 2009;15(1):100–109. [Europe PMC free article] [Abstract] [Google Scholar]
163. Kufe DW. Functional targeting of the MUC1 oncogene in human cancers. Cancer Biol Ther. 2009;8(13):1197–1203. [Europe PMC free article] [Abstract] [Google Scholar]
164. Raina D, Ahmad R, Joshi MD, et al. Direct targeting of the mucin 1 oncoprotein blocks survival and tumorigenicity of human breast carcinoma cells. Cancer Res. 2009;69(12):5133–5141. [Europe PMC free article] [Abstract] [Google Scholar]
165. Joshi MD, Ahmad R, Yin L, et al. MUC1 oncoprotein is a druggable target in human prostate cancer cells. Mol Cancer Ther. 2009;8(11):3056–3065. [Europe PMC free article] [Abstract] [Google Scholar]
166. Zhou Y, Rajabi H, Kufe D. Mucin 1 C-terminal subunit oncoprotein is a target for small-molecule inhibitors. Mol Pharmacol. 2011;79(5):886–893. [Europe PMC free article] [Abstract] [Google Scholar]
167. Torres MP, Ponnusamy MP, Chakraborty S, et al. Effects of thymoquinone in the expression of mucin 4 in pancreatic cancer cells: implications for the development of novel cancer therapies. Mol Cancer Ther. 2010;9(5):1419–1431. [Europe PMC free article] [Abstract] [Google Scholar]
168. Sangha R, Butts C. L-BLP25: a peptide vaccine strategy in non small cell lung cancer. Clin Cancer Res. 2007;13(15 Pt 2):s4652–s4654. [Abstract] [Google Scholar]
169. Sangha R, North S. L-BLP25: a MUC1-targeted peptide vaccine therapy in prostate cancer. Expert Opin Biol Ther. 2007;7(11):1723–1730. [Abstract] [Google Scholar]
170. Ramlau R, Quoix E, Rolski J, et al. A Phase II study of Tg4010 (Mva-Muc1-Il2) in association with chemotherapy in patients with stage III/IV non-small cell lung cancer. J Thorac Oncol. 2008;3(7):735–744. [Abstract] [Google Scholar]
171. Kaufman HL, Kim-Schulze S, Manson K, et al. Poxvirus-based vaccine therapy for patients with advanced pancreatic cancer. J Transl Med. 2007;5:60. [Europe PMC free article] [Abstract] [Google Scholar]
172. Gulley JL, Arlen PM, Tsang KY, et al. Pilot study of vaccination with recombinant CEA-MUC-1-TRICOM poxviral-based vaccines in patients with metastatic carcinoma. Clin Cancer Res. 2008;14(10):3060–3069. [Europe PMC free article] [Abstract] [Google Scholar]
173. Tang CK, Katsara M, Apostolopoulos V. Strategies used for MUC1 immunotherapy: human clinical studies. Expert Rev Vaccines. 2008;7(7):963–975. [Abstract] [Google Scholar]
174. Savla R, Taratula O, Garbuzenko O, Minko T. Tumor targeted quantum dot-mucin 1 aptamer-doxorubicin conjugate for imaging and treatment of cancer. J Control Release. 2011;153(1):16–22. [Abstract] [Google Scholar]
175. Ferreira CS, Matthews CS, Missailidis S. DNA aptamers that bind to MUC1 tumour marker: design and characterization of MUC1-binding single-stranded DNA aptamers. Tumour Biol. 2006;27(6):289–301. [Abstract] [Google Scholar]
176. Glazer ES, Zhu C, Massey KL, et al. Noninvasive radiofrequency field destruction of pancreatic adenocarcinoma xenografts treated with targeted gold nanoparticles. Clin Cancer Res. 2010;16(23):5712–5721. [Europe PMC free article] [Abstract] [Google Scholar]
177. Constantinou PE, Danysh BP, Lukianova-Helb EY, Lapotko DO, Carson DD. Targeted nanoparticle delivery and cytolysis. Presented at: Nano in Cancer: Linking Chemistry, Biology, and Clinical Applications In Vivo (AACR Special Conference); Miami, FL, USA. 12–15 January 2011. [Google Scholar]

Websites

201. National Institutes of Health Registry of Clinical Trials. www.clinicaltrials.gov.
202. Press release: Therion Reports Results Of Phase 3 PANVAC-VF Trial And Announces Plans For Company Sale. www.medicalnewstoday.com/releases/46137.php.

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Article citations


Go to all (19) article citations

Funding 


Funders who supported this work.

NCI NIH HHS (4)

NICHD NIH HHS (2)