Next Article in Journal
Gut Barrier Damage and Gut Translocation of Pathogen Molecules in Lupus, an Impact of Innate Immunity (Macrophages and Neutrophils) in Autoimmune Disease
Next Article in Special Issue
Inhibition of the Phospholipase Cε–c-Jun N-Terminal Kinase Axis Suppresses Glioma Stem Cell Properties
Previous Article in Journal
Circadian Rhythm Dysregulation and Leukemia Development: The Role of Clock Genes as Promising Biomarkers
Previous Article in Special Issue
HDAC Class I Inhibitor Domatinostat Preferentially Targets Glioma Stem Cells over Their Differentiated Progeny
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Addiction of Cancer Stem Cells to MUC1-C in Triple-Negative Breast Cancer Progression

Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(15), 8219; https://doi.org/10.3390/ijms23158219
Submission received: 12 July 2022 / Revised: 20 July 2022 / Accepted: 21 July 2022 / Published: 26 July 2022
(This article belongs to the Special Issue Cellular and Molecular Biology of Cancer Stem Cells)

Abstract

:
Triple-negative breast cancer (TNBC) is an aggressive malignancy with limited treatment options. TNBC progression is associated with expansion of cancer stem cells (CSCs). Few insights are available regarding druggable targets that drive the TNBC CSC state. This review summarizes the literature on TNBC CSCs and the compelling evidence that they are addicted to the MUC1-C transmembrane protein. In normal epithelia, MUC1-C is activated by loss of homeostasis and induces reversible wound-healing responses of inflammation and repair. However, in settings of chronic inflammation, MUC1-C promotes carcinogenesis. MUC1-C induces EMT, epigenetic reprogramming and chromatin remodeling in TNBC CSCs, which are dependent on MUC1-C for self-renewal and tumorigenicity. MUC1-C-induced lineage plasticity in TNBC CSCs confers DNA damage resistance and immune evasion by chronic activation of inflammatory pathways and global changes in chromatin architecture. Of therapeutic significance, an antibody generated against the MUC1-C extracellular domain has been advanced in a clinical trial of anti-MUC1-C CAR T cells and in IND-enabling studies for development as an antibody–drug conjugate (ADC). Agents targeting the MUC1-C cytoplasmic domain have also entered the clinic and are undergoing further development as candidates for advancing TNBC treatment. Eliminating TNBC CSCs will be necessary for curing this recalcitrant cancer and MUC1-C represents a promising druggable target for achieving that goal.

1. Background

1.1. Challenges in the Treatment of Advanced Triple-Negative Breast Cancer (TNBC)

Metastatic TNBC typically has a highly aggressive clinical course [1,2]. Cytotoxic chemotherapy remains the standard treatment for advanced disease [1]. However, in the metastatic setting, patients treated with platinum-based regimens have limited increases in progression-free survival as compared to those receiving other available options [3]. Germline BRCA1/2 gene mutations, which occur in approximately 10% of patients, increase responsiveness to platinum-based agents [4]. In addition, TNBCs with biallelic BRCA1/2 loss are dependent on PARP-mediated DNA repair and are sensitive to PARP inhibitors [5]. Despite these advances, patients invariably develop resistance to these agents and succumb to progressive disease [2].
Treatment of advanced TNBC with immune checkpoint inhibitors (ICIs) as monotherapy has also been associated with limited clinical benefit. Patients with PD-L1+ TNBCs and fewer previous lines of chemotherapy are more likely to respond to ICIs [2]. However, intrinsic resistance to ICIs occurs in 60–85% of patients [2]. Moreover, most patients who experience stable disease or objective responses develop acquired ICI resistance and few have lasting benefits [2]. Combining ICIs with chemotherapy has had more promising activity. Pembrolizumab in combination with chemotherapy has been approved as a first-line treatment for PD-L1+ advanced TNBC [6,7,8,9,10]. ICIs are also being combined with PARP inhibitors and antibody–drug conjugates (ADCs) for evaluation in clinical studies [2,11,12,13].
Major obstacles for the treatment of advanced TNBC are thus intrinsic and acquired resistance to genotoxic and immunotherapeutic agents. The cancer stem cell (CSC) state is associated with lineage plasticity, DNA damage resistance and immune evasion [14,15,16,17], indicating that elimination of CSCs will be necessary for achieving durable responses and cures. The effective treatment of TNBC will therefore clearly require the identification of druggable targets that are essential for the CSC state.

1.2. Potential Therapeutic Targets of the TNBC CSC State

Recent articles have reviewed the importance of CSCs in TNBC progression and therapeutic resistance [18,19,20,21,22,23]. TNBCs are enriched with CD44+/CD24−/ALDH1+ CSCs that contribute to metastases, chemoresistance and poor clinical outcomes [24,25,26,27,28]. Resistance of TNBC CSCs to DNA damage is mediated by phenotypic heterogeneity and epithelial–mesenchymal plasticity, which are driven in part by the ZEB1 transcription factor (TF) [29,30,31,32]. The WNT, NF-κB and NOTCH pathways are activated in TNBC CSCs [20,33]. Activation of Sonic Hedgehog (SHH) has also been identified as a pathway that contributes to the TNBC CSC state [33,34,35]. Agents that target WNT, NF-κB, NOTCH and SHH signaling are under development to eliminate CSCs in TNBCs, as well as other cancers [20,21]. Along these lines, small molecule inhibitors of WNT/β-catenin/TCF signaling are being developed based on their inhibition of TNBC CSCs in cell line and tumor models [36,37,38]. Antibodies that inhibit the NOTCH pathway by blocking gamma-secretase-mediated cleavage and receptor–ligand interactions represent potential CSC-directed therapeutics [39]. In addition, a GLI inhibitor has been under investigation for targeting the SHH pathway in CSCs [40]. As another approach, a genomewide RNAi screen was used to identify potential therapeutic targets that control the BC CSC state, and this work led to the identification of salinomycin and JQ1 as agents with activity in TNBC PDX models [41]. Nonetheless, to our knowledge, these antibodies and signaling pathway inhibitors yet been advanced clinically as effective TNBC therapeutics.
Trophoblast cell surface antigen-2 (TROP-2) is a transmembrane glycoprotein that is overexpressed by TNBCs and other types of cancers [42,43]. TROP-2 plays a role in the self-renewal of stem cells and thus represents a potential target for TNBC CSCs [42,43]. Sacituzumab govitecan (SG; IMMU-1320) is an ADC that targets TROP-2 for the selective delivery of the irinotecan metabolite SN-38 [44]. SG is the first ADC approved for the treatment of TNBC based on activity in the advanced-disease setting [45,46,47,48]. In contrast to MMAE, which targets microtubules with cell cycle arrest, SN-38 is a topoisomerase I inhibitor that induces DNA damage [44]. The efficacy of SG is attributed to direct targeting of TROP-2-expressing TNBC cells, as well as the release of SN-38 into the TME with the killing of nontargeted cells [44].
Other approaches that inhibit signaling pathways shared by TNBC CSCs and non-CSCs, including PARP and CDK4/6 inhibitors [2,49], are under active development; however, what is needed, at least in part, are agents that selectively target TNBC CSCs. The present review summarizes compelling evidence that TNBC CSCs are addicted to the MUC1-C oncogenic protein, against which agents are under clinical and preclinical evaluation for TNBC treatment.

1.3. Discovery of Mucin 1 (MUC1)

A monoclonal antibody, designated MAb DF3, was generated against a membrane-enriched fraction from a human breast carcinoma [50]. Studies with MAb DF3 led to the identification of the DF3 high molecular weight (>300 kD) protein. DF3 was found to be overexpressed on the entire cell surface and in the cytoplasm of breast cancer cells [50]. In contrast, DF3 was detectable at lower levels at the apical borders of normal breast epithelial cells [50]. MAbs generated against human milk fat globules (HMFG-1 and HMFG-2) were similarly found to recognize what was identified as the high molecular weight polymorphic epithelial membrane antigen (EMA, PEM) expressed on human breast cancer cells [51,52]. These early findings formed the basis for multiple studies of DF3 and PEM that explored their physical structures and expression patterns in normal and cancer cells. An advance in the field at that time was the discovery that the gene encoding these high molecular weight glycoproteins is frequently altered in breast cancers and contains conserved variable numbers of 60 base-pair tandem repeats (TRs) [53,54,55,56]. The encoded 20 amino acid TRs were found to be extensively modified by O-glycosylation, which identified a unique structure in a family of some 23 secreted and transmembrane mucins with the founding DF3/EMA/PEM member entitled mucin 1 (MUC1) [57]. These early findings collectively provided support for the potential importance of MUC1 as a cancer target [58]. Indeed, after a decade of fundamental and translational research, this progress in the field attracted interest in MUC1 as a potential target for developing anticancer vaccines and other therapeutic approaches [57,58].

1.4. Further Characterization of MUC1 as an Oncoprotein

MUC1 consists of two subunits that are formed by autoproteolysis of the translated protein [59]: (i) an extracellular N-terminal subunit (MUC1-N) with the glycosylated TRs that are characteristic of the mucin family and (ii) a transmembrane C-terminal subunit (MUC1-C) [57,58]. The MUC1-N/MUC1-C heterodimeric complex is expressed at the apical borders of normal epithelial cells, where it functions in protection against damage and inflammation (Figure 1) [57,58,60,61,62]. With loss of apical–basal polarity in response to disruption of homeostasis, MUC1-N is shed into a protective mucin barrier, where it forms complexes with pathogens that are then excreted by mucociliary transport. In addition, MUC1-C is expressed over the entire epithelial cell surface [58]. As a result, MUC1-C (i) interacts with receptor tyrosine kinases (RTKs) and other cell surface molecules normally positioned at the basal-lateral borders and (ii) integrates activation of cell membrane and intracellular signaling pathways (Figure 1).
MUC1-C consists of a 58-amino acid (aa) extracellular domain that is glycosylated on Asn-36, which then functions as a binding site for the galectin-3 ligand [63,64]. In turn, galectin-3 acts as a bridge to facilitate the association of MUC1-C with EGFR, FGFR, HER2 and other RTKs, and to activate their downstream signaling pathways (Figure 1) [63,64,65,66,67]. Activation of MUC1-C is associated with the formation of homodimers that are imported into the nucleus by importin-β and interactions with the nuclear pore complex [58,62,68]. In the nucleus, MUC1-C interacts directly with transcription factors, including STAT3 [69], WNT/β-catenin/TCF4 [70], NF-κB [71] and MYC [72], which as noted above have been linked to the CSC state, and contributes to the regulation of their target genes (Figure 1) [58,61]. MUC1-C is also transported by HSP70/HSP90 to the mitochondrial outer membrane, where it attenuates stress-induced apoptosis (Figure 1) [73,74].
The MUC1-C cytoplasmic domain consists of 72 aa, which include a CQC motif adjacent to the transmembrane domain that is necessary for the formation of homodimers and interactions with binding partners such as MYC [58,61,72]. Downstream of the CQC motif is an intrinsically disordered protein, as is often found in oncoproteins that integrate multiple signaling pathways [75,76].

1.5. MUC1-C Is of Importance to Hallmarks of TNBC Progression

MUC1-C contributes to HER2 activation and traztuzumab resistance in HER2+ breast cancer cells [67]. In addition, MUC1-C promotes tamoxifen resistance in ER+ breast cancer cells [77], indicating that it plays a significant role in different breast cancer subtypes. Here, we have focused on the importance of MUC1-C in the TNBC progression.
MUC1 is overexpressed in ~90% of TNBCs [58,61,78]. In concert with the capacity to intersect with diverse signaling pathways, MUC1-C is of importance to multiple hallmarks of breast cancer cells, including (i) increased growth [70], (ii) inhibition of death [79], (iii) metabolic alterations [80,81], (iv) induction of ZEB1, TWIST1 and EMT [82,83], (v) epigenetic reprogramming [84,85,86,87], (vi) chromatin remodeling [72,88], (vii) expression of stem cell markers [80,89] (viii) the CSC state, self-renewal capacity and tumorigenicity [83,90], (ix) genotoxic drug resistance [67,77,91,92,93] and (x) immune evasion [93,94,95].
Considerable evidence has emphasized the importance of lineage plasticity and the CSC state in driving therapeutic resistance [14,15,16,17]. In concert with that evidence, MUC1-C drives TNBC progression in association with induction of (i) EMT, (ii) self-renewal capacity, (iii) resistance to genotoxic agents and (iv) immune evasion [58]. These discoveries, which are summarized in more detail below, have collectively supported the addiction of TNBC CSCs to MUC1-C and have emphasized the importance of MUC1-C as a target for their elimination with agents developed against the MUC1-C extracellular and cytoplasmic domains.

1.6. TNBC CSCs Are Dependent on MUC1-C for Lineage Plasticity and Genotoxic Drug Resistance

MUC1-C interacts directly with the proinflammatory STAT3 and NF-κB TFs in cancer cells and contributes to the regulation of their target genes [58,61]. Involvement of MUC1-C in chronic activation of these pathways in cancer represents misappropriation of the role for MUC1-C in the protection of barrier epithelia from inflammation [57,58]. In TNBC cells, MUC1-C induces pSTAT3 and MUC1-C/pSTAT3 complexes that activate the TWIST1 gene [83]. MUC1-C also interacts with the TWIST1 EMT-TF in driving (i) MUC1 in an autoinductive circuit, (ii) ZEB1, SNAIL and other genes in the EMT program and (iii) the capacity for cell invasion. The MUC1-C/TWIST1 circuit also drives expression of the stem cell markers SOX2, BMI1, ALDH1 and CD44, self-renewal capacity and TNBC tumorigenicity. Consistent with the induction of lineage plasticity and the CSC state, MUC1-C and TWIST1 were shown to be necessary for acquired paclitaxel (PTX) resistance. MUC1-C drives luminal–basal dedifferentiation of TNBC cells by repressing ESR1 and downstream ER pathways in association with the downregulation of FOXA1, GATA3 and other markers of the luminal phenotype. MUC1-C also represses expression of BRCA1 [86], which plays a role in dictating mammary lineage stem cell fate [96]. In this way, MUC1-C integrates lineage plasticity, EMT and dedifferentiation in promoting the TNBC CSC state. Of translational relevance, targeting MUC1-C reverses PTX resistance as supported by synergistic activity with PTX against PTX-resistant TNBC cells. These findings uncovered dependence on MUC1-C for integrating lineage plasticity of TNBC CSCs with DNA damage resistance.
Mechanistic studies further demonstrated that MUC1-C activates ATM and promotes nuclear localization of the BMI1 and EZH2 polycomb group proteins, which form complexes with PARP1 in the DNA damage response (DDR) [92,97]. MUC1-C regulates (i) BMI1-induced H2A ubiquitylation, (ii) EZH2-driven H3K27 trimethylation and (iii) importantly, PARP1 activity. As a result, targeting MUC1-C sensitizes TNBC cells with mutant and wild-type BRCA1 to the PARP inhibitor olaparib. These findings (i) uncovered a role for MUC1-C in regulating PARP1 and promoting DNA damage resistance and (ii) identified a potential strategy for enhancing the effectiveness of taxanes, platinum-based agents and PARP inhibitors against TNBC CSCs.
Other mechanistic studies have demonstrated that MUC1-C promotes DNA damage resistance of TNBC CSCs by chronic activation of cytosolic nucleotide receptors and the cGAS-stimulator of IFN genes (STING) [93]. In TNBC CSCs, MUC1-C was found to be necessary for intrinsic expression of the RIG-I, MDA5 and cGAS cytosolic nucleotide pattern recognition receptors (PRRs) and STING. In concert with activating the PRR/STING axis, MUC1-C induces chronic IFN-β production and the inflammatory type I IFN pathway. As a result, MUC1-C induces the IFN-related DNA damage resistance gene signature (IRDS), which includes ISG15, in integrating chronic inflammation and DNA damage resistance. MUC1-C is also necessary for expression of PRRs, STING and ISG15 in the response of TNBC cells to treatment with carboplatin and olaparib, in further support of a critical role for MUC1-C in the DNA damage response (DDR). These findings and those with PTX demonstrated that MUC1-C plays a role in conferring intrinsic and acquired chemoresistance [98].

1.7. MUC1-C Promotes Immune Evasion of TNBC CSCs

The presence of tumor infiltrating lymphocytes (TILs), specifically CD8+ T cells, in the TNBC TME is associated with improved survival in response to chemotherapy [99,100,101]. The responsiveness of TNBCs to ICIs such as pembrolizumab is also improved by increases in TIL density [102,103,104,105,106], indicating that immune cell-depleted “cold” TMEs associate with resistance to treatment with genotoxic and immunotherapeutic agents. BRCA1/2 mutations and HR deficiency in TNBCs have not been associated with TIL density [107], indicating that factors other than mutational burden are of importance to depletion or dysfunction of TILs in the TNBC TME. CSCs have been linked to immune suppression across different types of cancer [15]. However, intrinsic effectors that contribute to the CSC state, “cold” TMEs and immune evasion remain largely known.
MUC1-C activates the PD-L1 gene in human TNBC cells by forming complexes with NF-κB on the PD-L1 promoter region [94]. Targeting MUC1-C in an immunocompetent TNBC GEMM in which mouse Eo771/MUC1 cells are engrafted onto human MUC1.Tg mice suppresses PD-L1 expression in association with increases in CD8+ T cells and tumor-cell killing. Moreover, MUC1 expression in human TNBC tumors correlates inversely with CD8+ T cells in the TME and survival, providing evidence that MUC1-C may contribute to TNBC immune evasion.
In support of those findings, subsequent studies demonstrated that MUC1-C activates the inflammatory type II IFN and JAK1→STAT1→IRF1 pathways in TNBC cells and tumors [95]. In this way, MUC1-C induces immunosuppressive effectors, including IDO1 and COX2/PTGS2, and associates with depletion and dysfunction of CD8+ T cells in the TNBC TME. These findings provided further evidence that MUC1-C activates immunosuppressive pathways in TNBCs and that MUC1-C is a potential target for inhibiting immune evasion of TNBC CSCs [98].

1.8. MUC1-C Is Necessary for the Remodeling of Chromatin in Driving the TNBC CSC State

Chromatin remodeling is critical for inducing the CSC state, lineage plasticity and therapeutic resistance. In concert with the dependence of TNBC CSCs on MUC1-C for DNA damage resistance and immune evasion, studies were performed to assess the effects of MUC1-C on reprogramming of chromatin to promote the CSC state. A previously unrecognized MUC1-C→MYC pathway was uncovered that regulates the NuRD chromatin remodeling and deacetylation complex [72]. A surprising discovery was that the MUC1-C cytoplasmic domain binds directly to the MYC HLH-LZ region, which also interacts with MAX. In turn, MUC1-C regulates the MYC transactivation function, which has opened a new area of investigation into involvement of MUC1-C in MYC signaling pathways. As one example, MUC1-C/MYC complexes activate the NuRD MTA1 and MBD3 genes in basal- and not luminal-type BC cells. In turn, MUC1-C recruits MTA1 and MBD3, as well as CHD4, to the ESR1 promoter, represses ESR1 expression and attenuates downstream ER signaling. This MUC1-C→MYC→NuRD pathway also suppressed expression of FOXA1, GATA3 and other effectors of the luminal phenotype in driving dedifferentiation of TNBC CSCs.
Other confirmatory work demonstrated that MUC1-C drives global changes in the chromatin architecture of TNBC CSCs [88]. MUC1-C induces differentially accessible regions (DARs) associated with differentially expressed genes (DEGs). Among these genes, we identified NOTCH1, which is required for TNBC cell self-renewal and the CSC state. MUC1-C activated NOTCH1 by recruiting JUN/AP-1 and the BAF chromatin remodeling complex to a proximal enhancer-like signature in association with increases in chromatin accessibility. Similar results were obtained for the EGR1 and LY6E stemness-associated genes, indicating that this MUC1-C-induced pathway of chromatin remodeling activates multiple genes that contribute to the TNBC CSC state. Taken together with regulation of the NuRD complex, these findings support an important role for MUC1-C in chromatin remodeling that drives luminal→basal dedifferentiation and lineage plasticity of TNBC CSCs [62].
These effects of MUC1-C on driving the TNBC CSC state extend to certain other cancers. In this regard, MUC1-C induces the Yamanaka pluripotency factors, EMT and self-renewal in pancreatic cancers [108] and castration-resistant prostate cancer (CRPC) [109]. In further support of the TNBC findings and consistent with driving the CSC state, MUC1-C also integrates lineage plasticity with chromatin remodeling, genotoxic drug resistance and immune evasion in these models [62,88,110,111,112].

1.9. Targeting MUC1-N for TNBC Treatment

Early attempts at targeting MUC1, particularly with antibodies, focused on MUC1-N and were ineffective in part because this subunit is shed from the cell surface [57,58,60,61]. In addition, the activity of antibodies generated against MUC1-N were limited by the large pool of circulating MUC1-N that has to be overcome for antibodies to reach the surface of cancer cells [58].
The provocative findings that the MUC1-N TRs are aberrantly under- or unglycosylated in cancer cells as compared to that in normal epithelia [113,114] invoked the possibility that vaccines and other immunotherapeutic approaches directed against the TRs could be developed to selectively treat human cancers. Indeed, after a decade of earlier work in the MUC1 field, groups of investigators became interested in studying preclinical and clinical anticancer vaccines for inducing immunity against unglycosylated TR peptides [115]. Despite this interest and the generation of anti-MUC1 TR immune responses, limited clinical benefit was observed in patients with cancer for advancing these vaccine approaches [116]. A notable example in this regard was the administration of the L-BLP25/Stimuvax/tecemotide vaccine, which induced an immune response against the MUC1-N TRs but was ineffective in prolonging overall survival in combination with chemoradiotherapy for patients with unresectable stage III NSCLC [58,117,118].
Aberrant O-glycosylation of the MUC1-N TRs in cancer results in truncated Tn and sialyl-Tn glycoforms that represent potential neoantigens for development of other immunotherapeutics [119]. Along these lines, anti-Tn-MUC1-N CAR-T cells developed by Dr. Carl June have entered a Phase 1 CART-TnMUC1-01 trial, which is now underway for therapy for patients with TnMUC1-positive advanced cancers (Tmunity Therapeutics).

1.10. MUC1-C Is a Target for Eliminating TNBC CSCs and Achieving Cures

In contrast to MUC1-N, the transmembrane MUC1-C subunit functions as an oncoprotein in TNBC CSC progression and is a potential druggable target for TNBC treatment [57,58,60,61,86,114]. A focus for advancing TNBC therapies has been the development of agents that target the MUC1-C 58 aa extracellular and 72 aa cytoplasmic domains [58].
MAb 3D1 was generated against the MUC1-C extracellular domain α-3 helix, which becomes exposed with MUC1-C activation [120] (Figure 2). Other antibodies are under development that target the junction between MUC1-N and MUC1-C [121,122] and the MUC1-C extracellular domain, including the SKM1 series [123] and MIN-C2 (Minerva Biotechnologies, Waltham, MA, USA). However, to our knowledge, there are no reports of MAbs that react with the α-3 helix.
The generation of MAb 3D1 against the MUC1-C extracellular domain provided an opportunity for the development of therapeutic agents, such as CAR T cells and ADCs, that target MUC1-C on the TNBC cell surface [120]. Along these lines, an anti-MUC1-C CAR T cell approach using MAb 3D1 sequences was highly effective against MDA-MB-468 TNBC tumor xenografts and has entered the clinic in a partnership with pharma for the treatment of TNBCs and other MUC1-C-expressing cancers (NCT05239143: P-MUC1C-ALLO1 Allogeneic CAR-T Cells in the Treatment of Subjects with Advanced or Metastatic Solid Tumors).
For the development of an ADC, MAb 3D1 was conjugated using a maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl (vc) cleavable linker to the toxin monomethyl auristatin E (MMAE). The vc cleavable linker is widely used with MMAE in approved ADCs, such as Adcetris (Seattle Genetics, Bothell, WA, USA), Polivy (Roche Genentech, South San Francisco, CA, USA) and Padcev (Astellas, Tokyo, Japan), as well as in other ADCs under development [124,125,126].
MAb3D1-MMAE ADCs were highly potent in killing MUC1-C-expressing cancer cells in vitro with an IC50 in the nM range [115]. The MAb3D1-MMAE ADCs were also effective against established human tumor xenografts, including TNBC PDX models, without evidence of overt toxicity [120]. To confirm the absence of toxicity in NCG mice expressing mouse Muc1-C, toxicity studies were performed in human MUC1.Tg mice that express MUC1 at a level and pattern found in humans [127]. Studies in the MUC1.Tg model demonstrated no significant adverse effects of the MAb3D1-MMAE ADC on normal tissues or hematologic parameters [120]. These findings supported the notion that the MUC1-C α-3 helix is selectively exposed on the surface of carcinoma cells as compared to normal epithelia [120]. Based on this therapeutic selectivity, the anti-MUC1-C ADCs are now under development by the NCI NExT Program for IND, which is enabling studies and conducting early phase trials in patients with TNBCs.
Other approaches have been under development for targeting the MUC1-C cytoplasmic domain (CD) [58]. MUC1-C CD is an intrinsically disordered protein devoid of kinase activity that includes a CQCRRK motif, which is necessary for MUC1-C homodimerization and function as an oncoprotein [58]. In this way, the MUC1-C cytoplasmic domain is phosphorylated by multiple kinases, such as RTKs, SRC, ABL and GSK3β, that in turn regulate direct interactions between MUC1-C and downstream effectors, including the MYC, NF-κB, STAT3 and WNT/β-catenin/TCF4 TFs, linked to the CSC state (Figure 3) [58,61,62].
Accordingly, the cell-penetrating GO-203 inhibitor (D-amino acids: (R9-CQCRRKN)) was developed to block the CQC motif and MUC1-C activity (Figure 3). In multiple studies, GO-203 exhibited dose-dependent activity against MUC1-expressing human tumor xenograft models [59,122,123,124,125]. In addition, GO-203 treatment phenocopies the effects of targeting MUC1-C genetically in cancer cells, supporting the use of this agent to inhibit MUC1-C function [58,108,109,128,129,130,131,132,133].
GO-203 is not a substrate for ABC transporters that are often upregulated in CSCs [58]. Along these lines, GO-203 is effective in inhibiting self-renewal capacity and tumorigenicity of human TNBC CSCs [58,72,83,92,133]. Consistent with resistance of TNBC CSCs to DNA damage, GO-203 is highly effective in potentiating the activity of genotoxic anticancer agents [58]. In studies of the Eo771/MUC1 GEMM TNBC, treatment with GO-203 also resulted in (i) downregulation of PD-L1 expression, (ii) activation of CD8+ T cells in the TME, (iii) induction of TILs effective in killing Eo771/MUC1 tumor cells and (iv) inhibition of Eo771/MUC1 tumor growth [94]. This TNBC GEMM thus has the potential for informing advances in the immunotherapy of TNBCs with anti-MUC1-C agents against the extracellular and cytoplasmic domains and for combination studies with immune-based therapies.

1.11. TNBC CSCs Are Addicted to MUC1-C for Lineage Plasticity and Therapeutic Resistance

In summary, TNBC CSCs are addicted to MUC1-C based on the definition that they are dependent on MUC1-C for survival [134]. TNBC CSCs are also dependent on MUC1-C for induction of EMT, epigenetic reprogramming and chromatin remodeling that are necessary for lineage plasticity, DNA damage resistance and immune evasion. These findings are of significance in having highlighted a critical role for MUC1-C in TNBC CSC progression and having emphasized the importance of developing agents that target the MUC1-C extracellular and cytoplasmic domains for TNBC treatment.

Author Contributions

Wrote and revised the review N.Y. and D.K. All authors have read and agreed to the published version of the manuscript.

Funding

Research reported in this publication was supported by the National Cancer Institute of the National Institutes of Health under grant numbers CA097098 and CA233084 awarded to D.K.

Conflicts of Interest

N.Y. declares no potential conflicts of interest. D.K. has equity interests in Genus Oncology, Reata Pharmaceuticals and HillstreamBiopharma and is a paid consultant to Reata and CanBas.

References

  1. Bianchini, G.; Balko, J.M.; Mayer, I.A.; Sanders, M.E.; Gianni, L. Triple-negative breast cancer: Challenges and opportunities of a heterogeneous disease. Nat. Rev. Clin. Oncol. 2016, 13, 674–690. [Google Scholar] [CrossRef] [PubMed]
  2. Bianchini, G.; De Angelis, C.; Licata, L.; Gianni, L. Treatment landscape of triple-negative breast cancer—Expanded options, evolving needs. Nat. Rev. Clin. Oncol. 2022, 19, 91–113. [Google Scholar] [CrossRef] [PubMed]
  3. Pandy, J.G.P.; Balolong-Garcia, J.C.; Cruz-Ordinario, M.V.B.; Que, F.V.F. Triple negative breast cancer and platinum-based systemic treatment: A meta-analysis and systematic review. BMC Cancer 2019, 19, 1065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Brok, W.D.D.; Schrader, K.A.; Sun, S.; Tinker, A.V.; Zhao, E.Y.; Aparicio, S.; Gelmon, K.A. Homologous Recombination Deficiency in Breast Cancer: A Clinical Review. JCO Precis. Oncol. 2017, 1, 1–13. [Google Scholar] [CrossRef]
  5. Layman, R.M.; Arun, B. PARP Inhibitors in Triple-Negative Breast Cancer Including Those With BRCA Mutations. Cancer J. 2021, 27, 67–75. [Google Scholar] [CrossRef] [PubMed]
  6. Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Hegg, R.; Im, S.-A.; Shaw Wright, G.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2018, 379, 2108–2121. [Google Scholar] [CrossRef]
  7. Emens, L.; Adams, S.; Barrios, C.; Diéras, V.; Iwata, H.; Loi, S.; Rugo, H.; Schneeweiss, A.; Winer, E.; Patel, S.; et al. Corrigendum to ‘First-line atezolizumab plus nab-paclitaxel for unresectable, locally advanced, or metastatic triple-negative breast cancer: IMpassion130 final overall survival analysis’. Ann. Oncol. 2021, 32, 1650. [Google Scholar] [CrossRef]
  8. Cortes, J.; Cescon, D.W.; Rugo, H.S.; Nowecki, Z.; Im, S.-A.; Yusof, M.M.; Gallardo, C.; Lipatov, O.; Barrios, C.H.; Holgado, E.; et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): A randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet 2020, 396, 1817–1828. [Google Scholar] [CrossRef]
  9. Carlino, F.; Diana, A.; Piccolo, A.; Ventriglia, A.; Bruno, V.; De Santo, I.; Letizia, O.; De Vita, F.; Daniele, B.; Ciardiello, F.; et al. Immune-Based Therapy in Triple-Negative Breast Cancer: From Molecular Biology to Clinical Practice. Cancers 2022, 14, 2102. [Google Scholar] [CrossRef]
  10. Huppert, L.A.; Gumusay, O.; Rugo, H.S. Emerging treatment strategies for metastatic triple-negative breast cancer. Ther. Adv. Med. Oncol. 2022, 14, 17588359221086916. [Google Scholar] [CrossRef]
  11. Vinayak, S.; Tolaney, S.M.; Schwartzberg, L.; Mita, M.; McCann, G.; Tan, A.R.; Wahner-Hendrickson, A.E.; Forero, A.; Anders, C.; Wulf, G.M.; et al. Open-label Clinical Trial of Niraparib Combined With Pembrolizumab for Treatment of Advanced or Metastatic Triple-Negative Breast Cancer. JAMA Oncol. 2019, 5, 1132–1140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Domchek, S.M.; Postel-Vinay, S.; Im, S.-A.; Park, Y.H.; Delord, J.-P.; Italiano, A.; Alexandre, J.; You, B.; Bastian, S.; Krebs, M.G.; et al. Olaparib and durvalumab in patients with germline BRCA-mutated metastatic breast cancer (MEDIOLA): An open-label, multicentre, phase 1/2, basket study. Lancet Oncol. 2020, 21, 1155–1164. [Google Scholar] [CrossRef]
  13. Schmid, P.; Abraham, J.; Chan, S.; Wheatley, D.; Brunt, A.M.; Nemsadze, G.; Baird, R.D.; Park, Y.H.; Hall, P.S.; Perren, T.; et al. Capivasertib Plus Paclitaxel Versus Placebo Plus Paclitaxel As First-Line Therapy for Metastatic Triple-Negative Breast Cancer: The PAKT Trial. J. Clin. Oncol. 2020, 38, 423–433. [Google Scholar] [CrossRef] [PubMed]
  14. De Angelis, M.L.; Francescangeli, F.; La Torre, F.; Zeuner, A. Stem Cell Plasticity and Dormancy in the Development of Cancer Therapy Resistance. Front. Oncol. 2019, 9, 626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Miranda, A.; Hamilton, P.T.; Zhang, A.W.; Pattnaik, S.; Becht, E.; Mezheyeuski, A.; Bruun, J.; Micke, P.; de Reynies, A.; Nelson, B.H. Cancer stemness, intratumoral heterogeneity, and immune response across cancers. Proc. Natl. Acad. Sci. USA 2019, 116, 9020–9029. [Google Scholar] [CrossRef] [Green Version]
  16. Malta, T.M.; Sokolov, A.; Gentles, A.J.; Burzykowski, T.; Poisson, L.; Weinstein, J.N.; Kamińska, B.; Huelsken, J.; Omberg, L.; Gevaert, O.; et al. Machine Learning Identifies Stemness Features Associated with Oncogenic Dedifferentiation. Cell 2018, 173, 338–354.e15. [Google Scholar] [CrossRef] [Green Version]
  17. Quintanal-Villalonga, Á.; Chan, J.M.; Yu, H.A.; Pe’Er, D.; Sawyers, C.L.; Sen, T.; Rudin, C.M. Lineage plasticity in cancer: A shared pathway of therapeutic resistance. Nat. Rev. Clin. Oncol. 2020, 17, 360–371. [Google Scholar] [CrossRef]
  18. O’Conor, C.J.; Chen, T.; González, I.; Cao, D.; Peng, Y. Cancer stem cells in triple-negative breast cancer: A potential target and prognostic marker. Biomark. Med. 2018, 12, 813–820. [Google Scholar] [CrossRef]
  19. Park, S.-Y.; Choi, J.-H.; Nam, J.-S. Targeting Cancer Stem Cells in Triple-Negative Breast Cancer. Cancers 2019, 11, 965. [Google Scholar] [CrossRef] [Green Version]
  20. Ehmsen, S.; Ditzel, H.J. Signaling pathways essential for triple-negative breast cancer stem-like cells. Stem Cells 2021, 39, 133–143. [Google Scholar] [CrossRef]
  21. He, L.; Wick, N.; Germans, S.K.; Peng, Y. The Role of Breast Cancer Stem Cells in Chemoresistance and Metastasis in Triple-Negative Breast Cancer. Cancers 2021, 13, 6209. [Google Scholar] [CrossRef] [PubMed]
  22. Fultang, N.; Chakraborty, M.; Peethambaran, B. Regulation of cancer stem cells in triple negative breast cancer. Cancer Drug Resist 2021, 4, 321. [Google Scholar] [CrossRef] [PubMed]
  23. Zong, Y.; Pegram, M. Research advances and new challenges in overcoming triple-negative breast cancer. Cancer Drug Resist 2021, 4, 517. [Google Scholar] [CrossRef] [PubMed]
  24. Honeth, G.; Bendahl, P.-O.; Ringnér, M.; Saal, L.H.; Gruvberger-Saal, S.K.; Lövgren, K.; Grabau, D.; Fernö, M.; Borg, A.; Hegardt, C. The CD44+/CD24-phenotype is enriched in basal-like breast tumors. Breast Cancer Res. 2008, 10, R53. [Google Scholar] [CrossRef] [Green Version]
  25. Li, H.; Ma, F.; Wang, H.; Lin, C.; Fan, Y.; Zhang, X.; Qian, H.; Xu, B. Stem Cell Marker Aldehyde Dehydrogenase 1 (ALDH1)-Expressing Cells are Enriched in Triple-Negative Breast Cancer. Int. J. Biol. Markers 2013, 28, 357–364. [Google Scholar] [CrossRef]
  26. Ma, F.; Li, H.; Wang, H.; Shi, X.; Fan, Y.; Ding, X.; Lin, C.; Zhan, Q.; Qian, H.; Xu, B. Enriched CD44+/CD24− population drives the aggressive phenotypes presented in triple-negative breast cancer (TNBC). Cancer Lett. 2014, 353, 153–159. [Google Scholar] [CrossRef]
  27. Lawson, D.A.; Bhakta, N.R.; Kessenbrock, K.; Prummel, K.D.; Yu, Y.; Takai, K.; Zhou, A.; Eyob, H.; Balakrishnan, S.; Wang, C.-Y.; et al. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature 2015, 526, 131–135. [Google Scholar] [CrossRef] [PubMed]
  28. Moreira, M.P.; Da Conceição Braga, L.; Cassali, G.D.; Silva, L.M. STAT3 as a promising chemoresistance biomarker associated with the CD44 +/high /CD24 -/low /ALDH + BCSCs-like subset of the triple-negative breast cancer (TNBC) cell line. Exp. Cell Res. 2018, 363, 283–290. [Google Scholar] [CrossRef]
  29. Kvokačková, B.; Remšík, J.; Jolly, M.; Souček, K. Phenotypic Heterogeneity of Triple-Negative Breast Cancer Mediated by Epithelial–Mesenchymal Plasticity. Cancers 2021, 13, 2188. [Google Scholar] [CrossRef]
  30. Feldker, N.; Ferrazzi, F.; Schuhwerk, H.; Widholz, S.A.; Guenther, K.; Frisch, I.; Jakob, K.; Kleemann, J.; Riegel, D.; Bönisch, U.; et al. Genome-wide cooperation of EMT transcription factor ZEB 1 with YAP and AP -1 in breast cancer. EMBO J. 2020, 39, e103209. [Google Scholar] [CrossRef]
  31. Vijay, G.V.; Na Zhao, N.; Hollander, P.D.; Toneff, M.J.; Joseph, R.; Pietila, M.; Taube, J.H.; Sarkar, T.R.; Ramirez-Pena, E.; Werden, S.J.; et al. GSK3β regulates epithelial-mesenchymal transition and cancer stem cell properties in triple-negative breast cancer. Breast Cancer Res. 2019, 21, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Drápela, S.; Bouchal, J.; Jolly, M.K.; Culig, Z.; Souček, K. ZEB1: A Critical Regulator of Cell Plasticity, DNA Damage Response, and Therapy Resistance. Front. Mol. Biosci. 2020, 7, 36. [Google Scholar] [CrossRef] [PubMed]
  33. Rangel, M.C.; Bertolette, D.; Castro, N.P.; Klauzinska, M.; Cuttitta, F.; Salomon, D.S. Developmental signaling pathways regulating mammary stem cells and contributing to the etiology of triple-negative breast cancer. Breast Cancer Res. Treat. 2016, 156, 211–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Habib, J.G.; O’Shaughnessy, J.A. The hedgehog pathway in triple-negative breast cancer. Cancer Med. 2016, 5, 2989–3006. [Google Scholar] [CrossRef] [PubMed]
  35. Dittmer, J. Breast cancer stem cells: Features, key drivers and treatment options. Semin. Cancer Biol. 2018, 53, 59–74. [Google Scholar] [CrossRef]
  36. Sulaiman, A.; Sulaiman, B.; Khouri, L.; McGarry, S.; Nessim, C.; Arnaout, A.; Li, X.; Addison, C.; Dimitroulakos, J.; Wang, L. Both bulk and cancer stem cell subpopulations in triple-negative breast cancer are susceptible to Wnt, HDAC, and ERα coinhibition. FEBS Lett. 2016, 590, 4606–4616. [Google Scholar] [CrossRef]
  37. Krishnamurthy, N.; Kurzrock, R. Targeting the Wnt/beta-catenin pathway in cancer: Update on effectors and inhibitors. Cancer Treat. Rev. 2018, 62, 50–60. [Google Scholar] [CrossRef]
  38. Jang, G.-B.; Hong, I.-S.; Kim, R.-J.; Lee, S.-Y.; Park, S.-J.; Lee, E.-S.; Park, J.H.; Yun, C.-H.; Chung, J.-U.; Lee, K.-J.; et al. Wnt/β-Catenin Small-Molecule Inhibitor CWP232228 Preferentially Inhibits the Growth of Breast Cancer Stem-like Cells. Cancer Res. 2015, 75, 1691–1702. [Google Scholar] [CrossRef] [Green Version]
  39. Tamagnone, L.; Zacchigna, S.; Rehman, M. Taming the Notch Transcriptional Regulator for Cancer Therapy. Molecules 2018, 23, 431. [Google Scholar] [CrossRef] [Green Version]
  40. Infante, P.; Alfonsi, R.; Botta, B.; Mori, M.; Di Marcotullio, L. Targeting GLI factors to inhibit the Hedgehog pathway. Trends Pharmacol. Sci. 2015, 36, 547–558. [Google Scholar] [CrossRef] [Green Version]
  41. Arfaoui, A.; Rioualen, C.; Azzoni, V.; Pinna, G.; Finetti, P.; Wicinski, J.; Josselin, E.; Macario, M.; Castellano, R.; Léonard-Stumpf, C.; et al. A genome-wide RNA i screen reveals essential therapeutic targets of breast cancer stem cells. EMBO Mol. Med. 2019, 11, e9930. [Google Scholar] [CrossRef] [PubMed]
  42. Goldenberg, D.M.; Stein, R.; Sharkey, R.M. The emergence of trophoblast cell-surface antigen 2 (TROP-2) as a novel cancer target. Oncotarget 2018, 9, 28989–29006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Zaman, S.; Jadid, H.; Denson, A.C.; Gray, J.E. Targeting Trop-2 in solid tumors: Future prospects. OncoTargets Ther. 2019, 12, 1781–1790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Cardillo, T.M.; Govindan, S.V.; Sharkey, R.M.; Trisal, P.; Arrojo, R.; Liu, D.; Rossi, E.A.; Chang, C.-H.; Goldenberg, D.M. Sacituzumab Govitecan (IMMU-132), an Anti-Trop-2/SN-38 Antibody–Drug Conjugate: Characterization and Efficacy in Pancreatic, Gastric, and Other Cancers. Bioconjugate Chem. 2015, 26, 919–931. [Google Scholar] [CrossRef]
  45. Bardia, A.; Mayer, I.A.; Diamond, J.R.; Moroose, R.L.; Isakoff, S.J.; Starodub, A.N.; Shah, N.C.; O’Shaughnessy, J.; Kalinsky, K.; Guarino, M.; et al. Efficacy and Safety of Anti-Trop-2 Antibody Drug Conjugate Sacituzumab Govitecan (IMMU-132) in Heavily Pretreated Patients With Metastatic Triple-Negative Breast Cancer. J. Clin. Oncol. 2017, 35, 2141–2148. [Google Scholar] [CrossRef]
  46. Schreiber, A.R.; Andress, M.; Diamond, J.R. Tackling metastatic triple-negative breast cancer with sacituzumab govitecan. Expert Rev. Anticancer Ther. 2021, 21, 1303–1311. [Google Scholar] [CrossRef] [PubMed]
  47. Kaboli, P.J.; Imani, S.; Jomhori, M.; Ling, K.H. Chemoresistance in breast cancer: PI3K/Akt pathway inhibitors vs the current chemotherapy. Am. J. Cancer Res. 2021, 11, 5155–5183. [Google Scholar]
  48. Carey, L.A.; Loirat, D.; Punie, K.; Bardia, A.; Diéras, V.; Dalenc, F.; Diamond, J.R.; Fontaine, C.; Wang, G.; Rugo, H.S.; et al. Sacituzumab govitecan as second-line treatment for metastatic triple-negative breast cancer—Phase 3 ASCENT study subanalysis. npj Breast Cancer 2022, 8, 72. [Google Scholar] [CrossRef]
  49. Gupta, G.K.; Collier, A.L.; Lee, D.; Hoefer, R.A.; Zheleva, V.; Van Reesema, L.L.S.; Tang-Tan, A.M.; Guye, M.L.; Chang, D.Z.; Winston, J.S.; et al. Perspectives on Triple-Negative Breast Cancer: Current Treatment Strategies, Unmet Needs, and Potential Targets for Future Therapies. Cancers 2020, 12, 2392. [Google Scholar] [CrossRef]
  50. Kufe, D.; Inghirami, G.; Abe, M.; Hayes, D.; Justi-Wheeler, H.; Schlom, J. Differential Reactivity of a Novel Monoclonal Antibody (DF3) with Human Malignant versus Benign Breast Tumors. Hybridoma 1984, 3, 223–232. [Google Scholar] [CrossRef]
  51. Burchell, J.; Durbin, H.; Taylor-Papadimitriou, J. Complexity of expression of antigenic determinants, recognized by monoclonal antibodies HMFG-1 and HMFG-2, in normal and malignant human mammary epithelial cells. J. Immunol. 1983, 131, 508–513. [Google Scholar] [PubMed]
  52. Ormerod, M.G.; Steele, K.; A Edwards, P.; Taylor-Papadimitriou, J. Monoclonal antibodies that react with epithelial membrane antigen. J. Exp. Pathol. 1984, 1, 263–271. [Google Scholar] [PubMed]
  53. Siddiqui, J.; Abe, M.; Hayes, D.; Shani, E.; Yunis, E.; Kufe, D. Isolation and sequencing of a cDNA coding for the human DF3 breast carcinoma-associated antigen. Proc. Natl. Acad. Sci. USA 1988, 85, 2320–2323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Merlo, G.R.; Siddiqui, J.; Cropp, C.S.; Liscia, D.S.; Lidereau, R.; Callahan, R.; Kufe, D.W. Frequent alteration of the DF3 tumor-associated antigen gene in primary human breast carcinomas. Cancer Res. 1989, 49, 6966–6971. [Google Scholar]
  55. Gendler, S.; Taylor-Papadimitriou, J.; Duhig, T.; Rothbard, J.; Burchell, J. A highly immunogenic region of a human polymorphic epithelial mucin expressed by carcinomas is made up of tandem repeats. J. Biol. Chem. 1988, 263, 12820–12823. [Google Scholar] [CrossRef]
  56. Gendler, S.J.; A Lancaster, C.; Taylor-Papadimitriou, J.; Duhig, T.; Peat, N.; Burchell, J.; Pemberton, L.; Lalani, E.N.; Wilson, D. Molecular cloning and expression of human tumor-associated polymorphic epithelial mucin. J. Biol. Chem. 1990, 265, 15286–15293. [Google Scholar] [CrossRef]
  57. Kufe, D.W. Mucins in cancer: Function, prognosis and therapy. Nat. Cancer 2009, 9, 874–885. [Google Scholar] [CrossRef] [Green Version]
  58. Kufe, D.W. MUC1-C in chronic inflammation and carcinogenesis; emergence as a target for cancer treatment. Carcinogenesis 2020, 41, 1173–1183. [Google Scholar] [CrossRef]
  59. Macao, B.; A Johansson, D.G.; Hansson, G.C.; Härd, T. Autoproteolysis coupled to protein folding in the SEA domain of the membrane-bound MUC1 mucin. Nat. Struct. Mol. Biol. 2006, 13, 71–76. [Google Scholar] [CrossRef]
  60. Kufe, N.W. Functional targeting of the MUC1 oncogene in human cancers. Cancer Biol. Ther. 2009, 8, 1197–1203. [Google Scholar] [CrossRef] [Green Version]
  61. Kufe, D.W. MUC1-C oncoprotein as a target in breast cancer: Activation of signaling pathways and therapeutic approaches. Oncogene 2013, 32, 1073–1081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kufe, D.W. Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells. J. Cancer Metastasis Treat. 2022, 8, 12. [Google Scholar] [CrossRef] [PubMed]
  63. Ramasamy, S.; Duraisamy, S.; Barbashov, S.; Kawano, T.; Kharbanda, S.; Kufe, D. The MUC1 and Galectin-3 Oncoproteins Function in a MicroRNA-Dependent Regulatory Loop. Mol. Cell 2007, 27, 992–1004. [Google Scholar] [CrossRef] [Green Version]
  64. Merlin, J.; Stechly, L.; de Beaucé, S.; Monté, D.; Leteurtre, E.; van Seuningen, I.; Huet, G.; Pigny, P. Galectin-3 regulates MUC1 and EGFR cellular distribution and EGFR downstream pathways in pancreatic cancer cells. Oncogene 2011, 30, 2514–2525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Li, Q.; Kuwahara, H.; Yin, L.; Li, Y.; Ren, J.; Yu, W.-H.; Iii, K.L.C.; Kufe, D. The Epidermal Growth Factor Receptor Regulates Interaction of the Human DF3/MUC1 Carcinoma Antigen with c-Src and β-Catenin. J. Biol. Chem. 2001, 276, 35239–35242. [Google Scholar] [CrossRef] [Green Version]
  66. Ren, J.; Raina, D.; Chen, W.; Li, G.; Huang, L.; Kufe, D. MUC1 Oncoprotein Functions in Activation of Fibroblast Growth Factor Receptor Signaling. Mol. Cancer Res. 2006, 4, 873–883. [Google Scholar] [CrossRef] [Green Version]
  67. Raina, D.; Uchida, Y.; Kharbanda, A.; Rajabi, H.; Panchamoorthy, G.; Jin, C.; Scaltriti, M.; Baselga, J.; Kufe, D. Targeting the MUC1-C oncoprotein downregulates HER2 activation and abrogates trastuzumab resistance in breast cancer cells. Oncogene 2014, 33, 3422–3431. [Google Scholar] [CrossRef] [Green Version]
  68. Leng, Y.; Cao, C.; Ren, J.; Huang, L.; Chen, D.; Ito, M.; Kufe, D. Nuclear Import of the MUC1-C Oncoprotein Is Mediated by Nucleoporin Nup62. J. Biol. Chem. 2007, 282, 19321–19330. [Google Scholar] [CrossRef] [Green Version]
  69. Ahmad, R.; Rajabi, H.; Kosugi, M.; Joshi, M.D.; Alam, M.; Vasir, B.; Kawano, T.; Kharbanda, S.; Kufe, D. MUC1-C Oncoprotein Promotes STAT3 Activation in an Autoinductive Regulatory Loop. Sci. Signal. 2011, 4, ra9. [Google Scholar] [CrossRef] [Green Version]
  70. Rajabi, H.; Ahmad, R.; Jin, C.; Kosugi, M.; Alam, M.; Joshi, M.D.; Kufe, D. MUC1-C Oncoprotein Induces TCF7L2 Transcription Factor Activation and Promotes Cyclin D1 Expression in Human Breast Cancer Cells. J. Biol. Chem. 2012, 287, 10703–10713. [Google Scholar] [CrossRef] [Green Version]
  71. Ahmad, R.; Raina, D.; Joshi, M.D.; Kawano, T.; Kharbanda, S.; Kufe, D. MUC1-C oncoprotein functions as a direct activator of the NF-ĸB p65 transcription factor. Cancer Res. 2009, 69, 7013–7021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Hata, T.; Rajabi, H.; Takahashi, H.; Yasumizu, Y.; Li, W.; Jin, C.; Long, M.D.; Hu, Q.; Liu, S.; Fushimi, A.; et al. MUC1-C Activates the NuRD Complex to Drive Dedifferentiation of Triple-Negative Breast Cancer Cells. Cancer Res. 2019, 79, 5711–5722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ren, J.; Agata, N.; Chen, D.; Li, Y.; Yu, W.-H.; Huang, L.; Raina, D.; Chen, W.; Kharbanda, S.; Kufe, D. Human MUC1 carcinoma-associated protein confers resistance to genotoxic anticancer agents. Cancer Cell 2004, 5, 163–175. [Google Scholar] [CrossRef] [Green Version]
  74. Ren, J.; Bharti, A.; Raina, D.; Chen, W.; Ahmad, R.; Kufe, D. MUC1 oncoprotein is targeted to mitochondria by heregulin-induced activation of c-Src and the molecular chaperone HSP90. Oncogene 2006, 25, 20–31. [Google Scholar] [CrossRef] [Green Version]
  75. Raina, D.; Agarwal, P.; Lee, J.; Bharti, A.; McKnight, C.J.; Sharma, P.; Kharbanda, S.; Kufe, N. Characterization of the MUC1-C Cytoplasmic Domain as a Cancer Target. PLoS ONE 2015, 10, e0135156. [Google Scholar] [CrossRef] [PubMed]
  76. Uversky, V.N. Intrinsically disordered proteins and novel strategies for drug discovery. Expert Opin. Drug Discov. 2012, 7, 475–488. [Google Scholar] [CrossRef] [PubMed]
  77. Kharbanda, A.; Rajabi, H.; Jin, C.; Raina, D.; Kufe, D. Oncogenic MUC1-C Promotes Tamoxifen Resistance in Human Breast Cancer. Mol. Cancer Res. 2013, 11, 714–723. [Google Scholar] [CrossRef] [Green Version]
  78. Siroy, A.; Abdul-Karim, F.W.; Miedler, J.; Fong, N.; Fu, P.; Gilmore, H.; Baar, J. MUC1 is expressed at high frequency in early-stage basal-like triple-negative breast cancer. Hum. Pathol. 2013, 44, 2159–2166. [Google Scholar] [CrossRef] [Green Version]
  79. Ahmad, R.; Alam, M.; Rajabi, H.; Kufe, D. The MUC1-C Oncoprotein Binds to the BH3 Domain of the Pro-apoptotic BAX Protein and Blocks BAX Function. J. Biol. Chem. 2012, 287, 20866–20875. [Google Scholar] [CrossRef] [Green Version]
  80. Hasegawa, M.; Takahashi, H.; Rajabi, H.; Alam, M.; Suzuki, Y.; Yin, L.; Tagde, A.; Maeda, T.; Hiraki, M.; Sukhatme, V.P.; et al. Functional interactions of the cystine/glutamate antiporter, CD44v and MUC1-C oncoprotein in triple-negative breast cancer cells. Oncotarget 2016, 7, 11756–11769. [Google Scholar] [CrossRef] [Green Version]
  81. Kosugi, M.; Ahmad, R.; Alam, M.; Uchida, Y.; Kufe, N. MUC1-C Oncoprotein Regulates Glycolysis and Pyruvate Kinase m2 Activity in Cancer Cells. PLoS ONE 2011, 6, e28234. [Google Scholar] [CrossRef] [PubMed]
  82. Rajabi, H.; Alam, M.; Takahashi, H.; Kharbanda, A.; Guha, M.; Ahmad, R.; Kufe, D. MUC1-C oncoprotein activates the ZEB1/miR-200c regulatory loop and epithelial–mesenchymal transition. Oncogene 2014, 33, 1680–1689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hata, T.; Rajabi, H.; Yamamoto, M.; Jin, C.; Ahmad, R.; Zhang, Y.; Kui, L.; Li, W.; Yasumizu, Y.; Hong, D.; et al. Targeting MUC1-C Inhibits TWIST1 Signaling in Triple-Negative Breast Cancer. Mol. Cancer Ther. 2019, 18, 1744–1754. [Google Scholar] [CrossRef] [Green Version]
  84. Rajabi, H.; Tagde, A.; Alam, M.; Bouillez, A.; Pitroda, S.; Suzuki, Y.; Kufe, D. DNA methylation by DNMT1 and DNMT3b methyltransferases is driven by the MUC1-C oncoprotein in human carcinoma cells. Oncogene 2016, 35, 6439–6445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Hiraki, M.; Maeda, T.; Bouillez, A.; Alam, M.; Tagde, A.; Hinohara, K.; Suzuki, Y.; Markert, T.; Miyo, M.; Komura, K.; et al. MUC1-C activates BMI1 in human cancer cells. Oncogene 2017, 36, 2791–2801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Rajabi, H.; Hiraki, M.; Tagde, A.; Alam, M.; Bouillez, A.; Christensen, C.L.; Samur, M.; Wong, K.-K.; Kufe, D. MUC1-C activates EZH2 expression and function in human cancer cells. Sci. Rep. 2017, 7, 7481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Rajabi, H.; Hiraki, M.; Kufe, D. MUC1-C activates polycomb repressive complexes and downregulates tumor suppressor genes in human cancer cells. Oncogene 2018, 37, 2079–2088. [Google Scholar] [CrossRef]
  88. Bhattacharya, A.; Fushimi, A.; Yamashita, N.; Hagiwara, M.; Morimoto, Y.; Rajabi, H.; Long, M.D.; Abdulla, M.; Ahmad, R.; Street, K.; et al. MUC1-C Dictates JUN and BAF-Mediated Chromatin Remodeling at Enhancer Signatures in Cancer Stem Cells. Mol. Cancer Res. 2022, 20, 556–567. [Google Scholar] [CrossRef]
  89. Alam, M.; Ahmad, R.; Rajabi, H.; Kharbanda, A.; Kufe, D. MUC1-C Oncoprotein Activates ERK→C/EBPβ Signaling and Induction of Aldehyde Dehydrogenase 1A1 in Breast Cancer Cells. J. Biol. Chem. 2013, 288, 30892–30903. [Google Scholar] [CrossRef] [Green Version]
  90. Alam, M.; Rajabi, H.; Ahmad, R.; Jin, C.; Kufe, D. Targeting the MUC1-C oncoprotein inhibits self-renewal capacity of breast cancer cells. Oncotarget 2014, 5, 2622–2634. [Google Scholar] [CrossRef] [Green Version]
  91. Uchida, Y.; Raina, D.; Kharbanda, S.; Kufe, D. Inhibition of the MUC1-C oncoprotein is synergistic with cytotoxic agents in the treatment of breast cancer cells. Cancer Biol. Ther. 2013, 14, 127–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Yamamoto, M.; Jin, C.; Hata, T.; Yasumizu, Y.; Zhang, Y.; Hong, D.; Maeda, T.; Miyo, M.; Hiraki, M.; Suzuki, Y.; et al. MUC1-C Integrates Chromatin Remodeling and PARP1 Activity in the DNA Damage Response of Triple-Negative Breast Cancer Cells. Cancer Res. 2019, 79, 2031–2041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Yamashita, N.; Fushimi, A.; Morimoto, Y.; Bhattacharya, A.; Hagiwara, M.; Yamamoto, M.; Hata, T.; Shapiro, G.I.; Long, M.D.; Liu, S.; et al. Targeting MUC1-C Suppresses Chronic Activation of Cytosolic Nucleotide Receptors and STING in Triple-Negative Breast Cancer. Cancers 2022, 14, 2580. [Google Scholar] [CrossRef]
  94. Maeda, T.; Hiraki, M.; Jin, C.; Rajabi, H.; Tagde, A.; Alam, M.; Bouillez, A.; Hu, X.; Suzuki, Y.; Miyo, M.; et al. MUC1-C Induces PD-L1 and Immune Evasion in Triple-Negative Breast Cancer. Cancer Res. 2018, 78, 205–215. [Google Scholar] [CrossRef] [Green Version]
  95. Yamashita, N.; Long, M.; Fushimi, A.; Yamamoto, M.; Hata, T.; Hagiwara, M.; Bhattacharya, A.; Hu, Q.; Wong, K.-K.; Liu, S.; et al. MUC1-C integrates activation of the IFN-γ pathway with suppression of the tumor immune microenvironment in triple-negative breast cancer. J. Immunother. Cancer 2021, 9, e002115. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, S.; Ginestier, C.; Charafe-Jauffret, E.; Foco, H.; Kleer, C.G.; Merajver, S.D.; Dontu, G.; Wicha, M.S. BRCA1 regulates human mammary stem/progenitor cell fate. Proc. Natl. Acad. Sci. USA 2008, 105, 1680–1685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Huang, L.; Liao, X.; Beckett, M.; Li, Y.; Khanna, K.K.; Wang, Z.; Kharbanda, S.; Weichselbaum, R.; Kufe, D. MUC1-C Oncoprotein Interacts Directly with ATM and Promotes the DNA Damage Response to Ionizing Radiation. Genes Cancer 2010, 1, 239–250. [Google Scholar] [CrossRef] [PubMed]
  98. Rajabi, H.; Kufe, D. MUC1-C Oncoprotein Integrates a Program of EMT, Epigenetic Reprogramming and Immune Evasion in Human Carcinomas. Biochim. Biophys. Acta (BBA) Rev. Cancer 2017, 1868, 117–122. [Google Scholar] [CrossRef]
  99. Loi, S.; Drubay, D.; Adams, S.; Pruneri, G.; Francis, P.A.; Lacroix-Triki, M.; Joensuu, H.; Dieci, M.V.; Badve, S.; Demaria, S.; et al. Tumor-Infiltrating Lymphocytes and Prognosis: A Pooled Individual Patient Analysis of Early-Stage Triple-Negative Breast Cancers. J. Clin. Oncol. 2019, 37, 559–569. [Google Scholar] [CrossRef]
  100. Hida, A.I.; Watanabe, T.; Sagara, Y.; Kashiwaba, M.; Sagara, Y.; Aogi, K.; Ohi, Y.; Tanimoto, A. Diffuse distribution of tumor-infiltrating lymphocytes is a marker for better prognosis and chemotherapeutic effect in triple-negative breast cancer. Breast Cancer Res. Treat. 2019, 178, 283–294. [Google Scholar] [CrossRef]
  101. Park, J.H.; Jonas, S.F.; Bataillon, G.; Criscitiello, C.; Salgado, R.; Loi, S.; Viale, G.; Lee, H.J.; Dieci, M.V.; Kim, S.-B.; et al. Prognostic value of tumor-infiltrating lymphocytes in patients with early-stage triple-negative breast cancers (TNBC) who did not receive adjuvant chemotherapy. Ann. Oncol. 2019, 30, 1941–1949. [Google Scholar] [CrossRef] [PubMed]
  102. El Bairi, K.; Haynes, H.R.; Blackley, E.; Fineberg, S.; Shear, J.; Turner, S.; de Freitas, J.R.; Sur, D.; Amendola, L.C.; Gharib, M.; et al. The tale of TILs in breast cancer: A report from The International Immuno-Oncology Biomarker Working Group. npj Breast Cancer 2021, 7, 150. [Google Scholar] [CrossRef]
  103. Schmid, P.; Rugo, H.S.; Adams, S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Henschel, V.; Molinero, L.; Chui, S.Y.; et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): Updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2020, 21, 44–59. [Google Scholar] [CrossRef]
  104. Qureshi, S.; Chan, N.; George, M.; Ganesan, S.; Toppmeyer, D.; Omene, C. Immune Checkpoint Inhibitors in Triple Negative Breast Cancer: The Search for the Optimal Biomarker. Biomark. Insights 2022, 17, 11772719221078774. [Google Scholar] [CrossRef] [PubMed]
  105. Thomas, R.; Al-Khadairi, G.; Decock, J. Immune Checkpoint Inhibitors in Triple Negative Breast Cancer Treatment: Promising Future Prospects. Front. Oncol. 2020, 10, 600573. [Google Scholar] [CrossRef]
  106. Tarantino, P.; Corti, C.; Schmid, P.; Cortes, J.; Mittendorf, E.A.; Rugo, H.; Tolaney, S.M.; Bianchini, G.; Andrè, F.; Curigliano, G. Immunotherapy for early triple negative breast cancer: Research agenda for the next decade. npj Breast Cancer 2022, 8, 23. [Google Scholar] [CrossRef]
  107. Telli, M.L.; Chu, C.; Badve, S.S.; Vinayak, S.; Silver, D.P.; Isakoff, S.J.; Kaklamani, V.; Gradishar, W.; Stearns, V.; Connolly, R.M.; et al. Association of Tumor-Infiltrating Lymphocytes with Homologous Recombination Deficiency and BRCA1/2 Status in Patients with Early Triple-Negative Breast Cancer: A Pooled Analysis. Clin. Cancer Res. 2020, 26, 2704–2710. [Google Scholar] [CrossRef]
  108. Luan, Z.; Morimoto, Y.; Fushimi, A.; Yamashita, N.; Suo, W.; Bhattacharya, A.; Hagiwara, M.; Jin, C.; Kufe, D. MUC1-C dictates neuroendocrine lineage specification in pancreatic ductal adenocarcinomas. Carcinogenesis 2021, 43, 67–76. [Google Scholar] [CrossRef]
  109. Yasumizu, Y.; Rajabi, H.; Jin, C.; Hata, T.; Pitroda, S.; Long, M.D.; Hagiwara, M.; Li, W.; Hu, Q.; Liu, S.; et al. MUC1-C regulates lineage plasticity driving progression to neuroendocrine prostate cancer. Nat. Commun. 2020, 11, 338. [Google Scholar] [CrossRef] [Green Version]
  110. Hagiwara, M.; Yasumizu, Y.; Yamashita, N.; Rajabi, H.; Fushimi, A.; Long, M.D.; Li, W.; Bhattacharya, A.; Ahmad, R.; Oya, M.; et al. MUC1-C Activates the BAF (mSWI/SNF) Complex in Prostate Cancer Stem Cells. Cancer Res. 2021, 81, 1111–1122. [Google Scholar] [CrossRef]
  111. Hagiwara, M.; Fushimi, A.; Yamashita, N.; Bhattacharya, A.; Rajabi, H.; Long, M.D.; Yasumizu, Y.; Oya, M.; Liu, S.; Kufe, D. MUC1-C activates the PBAF chromatin remodeling complex in integrating redox balance with progression of human prostate cancer stem cells. Oncogene 2021, 40, 4930–4940. [Google Scholar] [CrossRef] [PubMed]
  112. Hagiwara, M.; Fushimi, A.; Bhattacharya, A.; Yamashita, N.; Morimoto, Y.; Oya, M.; Withers, H.G.; Hu, Q.; Liu, T.; Liu, S.; et al. MUC1-C integrates type II interferon and chromatin remodeling pathways in immunosuppression of prostate cancer. OncoImmunology 2022, 11, 2029298. [Google Scholar] [CrossRef] [PubMed]
  113. Girling, A.; Bartkova, J.; Burchell, J.; Gendler, S.; Gillett, C.; Taylor-Papadimitriou, J. A core protein epitope of the polymorphic epithelial mucin detected by the monoclonal antibody SM-3 is selectively exposed in a range of primary carcinomas. Int. J. Cancer 1989, 43, 1072–1076. [Google Scholar] [CrossRef] [PubMed]
  114. Perey, L.; Hayes, D.F.; Maimonis, P.; Abe, M.; O’Hara, C.; Kufe, D.W. Tumor selective reactivity of a monoclonal antibody prepared against a recombinant peptide derived from the DF3 human breast carcinoma-associated antigen. Cancer Res. 1992, 52, 2563–2568. [Google Scholar]
  115. Kimura, T.; Finn, O.J. MUC1 immunotherapy is here to stay. Expert Opin. Biol. Ther. 2013, 13, 35–49. [Google Scholar] [CrossRef]
  116. Taylor-Papadimitriou, J.; Burchell, J.M.; Graham, R.; Beatson, R. Latest developments in MUC1 immunotherapy. Biochem. Soc. Trans. 2018, 46, 659–668. [Google Scholar] [CrossRef] [Green Version]
  117. Butts, C.; Socinski, M.A.; Mitchell, P.L.; Thatcher, N.; Havel, L.; Krzakowski, M.; Nawrocki, S.; Ciuleanu, T.E.; Bosquée, L.; Trigo, J.M.; et al. Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START): A randomised, double-blind, phase 3 trial. Lancet Oncol. 2014, 15, 59–68. [Google Scholar] [CrossRef] [Green Version]
  118. Mitchell, P.; Thatcher, N.; Socinski, M.A.; Wasilewska-Tesluk, E.; Horwood, K.; Szczesna, A.; Martín, C.; Ragulin, Y.; Zukin, M.; Helwig, C.; et al. Tecemotide in unresectable stage III non-small-cell lung cancer in the phase III START study: Updated overall survival and biomarker analyses. Ann. Oncol. 2015, 26, 1134–1142. [Google Scholar] [CrossRef]
  119. Steentoft, C.; Migliorini, D.; King, T.R.; Mandel, U.; June, C.H.; Posey, J.A. Glycan-directed CAR-T cells. Glycobiology 2018, 28, 656–669. [Google Scholar] [CrossRef]
  120. Panchamoorthy, G.; Jin, C.; Raina, D.; Bharti, A.; Yamamoto, M.; Adeebge, D.; Zhao, Q.; Bronson, R.; Jiang, S.; Li, L.; et al. Targeting the human MUC1-C oncoprotein with an antibody-drug conjugate. JCI Insight 2018, 3, e99880. [Google Scholar] [CrossRef]
  121. Pichinuk, E.; Chalik, M.; Benhar, I.; Ginat-Koton, R.; Ziv, R.; Smorodinsky, N.I.; Haran, G.; Garbar, C.; Bensussan, A.; Meeker, A.; et al. In vivo anti-MUC1+ tumor activity and sequences of high-affinity anti-MUC1-SEA antibodies. Cancer Immunol. Immunother. 2020, 69, 1337–1352. [Google Scholar] [CrossRef] [PubMed]
  122. Wu, G.; Li, L.; Qiu, Y.; Sun, W.; Ren, T.; Lv, Y.; Liu, M.; Wang, X.; Tao, H.; Zhao, L.; et al. A novel humanized MUC1 antibody–drug conjugate for the treatment of trastuzumab-resistant breast cancer. Acta Biochim. Biophys. Sin. 2021, 53, 1625–1639. [Google Scholar] [CrossRef] [PubMed]
  123. Kim, T.J.; Koo, K.C. Current Status and Future Perspectives of Checkpoint Inhibitor Immunotherapy for Prostate Cancer: A Comprehensive Review. Int. J. Mol. Sci. 2020, 21, 5484. [Google Scholar] [CrossRef] [PubMed]
  124. Akaiwa, M.; Dugal-Tessier, J.; Mendelsohn, B.A. Antibody–Drug Conjugate Payloads; Study of Auristatin Derivatives. Chem. Pharm. Bull. 2020, 68, 201–211. [Google Scholar] [CrossRef] [Green Version]
  125. Joubert, N.; Beck, A.; Dumontet, C.; Denevault-Sabourin, C. Antibody–Drug Conjugates: The Last Decade. Pharmaceuticals 2020, 13, 245. [Google Scholar] [CrossRef]
  126. Drago, J.Z.; Modi, S.; Chandarlapaty, S. Unlocking the potential of antibody–drug conjugates for cancer therapy. Nat. Rev. Clin. Oncol. 2021, 18, 327–344. [Google Scholar] [CrossRef]
  127. Rowse, G.J.; Tempero, R.M.; VanLith, M.L.; A Hollingsworth, M.; Gendler, S.J. Tolerance and immunity to MUC1 in a human MUC1 transgenic murine model. Cancer Res. 1998, 58, 315–321. [Google Scholar]
  128. Raina, D.; Kosugi, M.; Ahmad, R.; Panchamoorthy, G.; Rajabi, H.; Alam, M.; Shimamura, T.; Shapiro, G.I.; Supko, J.; Kharbanda, S.; et al. Dependence on the MUC1-C Oncoprotein in Non–Small Cell Lung Cancer Cells. Mol. Cancer Ther. 2011, 10, 806–816. [Google Scholar] [CrossRef] [Green Version]
  129. Kharbanda, A.; Rajabi, H.; Jin, C.; Tchaicha, J.; Kikuchi, E.; Wong, K.-K.; Kufe, D. Targeting the Oncogenic MUC1-C Protein Inhibits Mutant EGFR-Mediated Signaling and Survival in Non–Small Cell Lung Cancer Cells. Clin. Cancer Res. 2014, 20, 5423–5434. [Google Scholar] [CrossRef] [Green Version]
  130. Bouillez, A.; Rajabi, H.; Pitroda, S.; Jin, C.; Alam, M.; Kharbanda, A.; Tagde, A.; Wong, K.-K.; Kufe, D. Inhibition of MUC1-C Suppresses MYC Expression and Attenuates Malignant Growth in KRAS Mutant Lung Adenocarcinomas. Cancer Res. 2016, 76, 1538–1548. [Google Scholar] [CrossRef] [Green Version]
  131. GongSun, X.; Zhao, Y.; Jiang, B.; Xin, Z.; Shi, M.; Song, L.; Qin, Q.; Wang, Q.; Liu, X. Inhibition of MUC1-C regulates metabolism by AKT pathway in esophageal squamous cell carcinoma. J. Cell. Physiol. 2019, 234, 12019–12028. [Google Scholar] [CrossRef] [Green Version]
  132. Zhao, Y.-Q.; Wu, T.; Wang, L.-F.; Yin, B.; Shi, M.; Jiang, B.; Gong-Sun, X.; Song, X.-M.; Liu, X.-Y. Targeting MUC1-C reverses the cisplatin resistance of esophageal squamous cell carcinoma in vitro and in vivo. Trans. Cancer Res. 2021, 10, 645–655. [Google Scholar] [CrossRef] [PubMed]
  133. Hasegawa, M.; Sinha, R.K.; Kumar, M.; Alam, M.; Yin, L.; Raina, D.; Kharbanda, A.; Panchamoorthy, G.; Gupta, D.; Singh, H.; et al. Intracellular Targeting of the Oncogenic MUC1-C Protein with a Novel GO-203 Nanoparticle Formulation. Clin. Cancer Res. 2015, 21, 2338–2347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Weinstein, I.B.; Joe, A. Oncogene Addiction. Cancer Res. 2008, 68, 3077–3080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. MUC1-C integrates activation of TNBC cell membrane signaling with the induction of EMT, lineage plasticity, DNA damage resistance and immune evasion. Activation of MUC1-C by loss of homeostasis is associated with the formation of homodimers that interact with EGFR and other RTKs at the cell membrane. MUC1-C homodimers are internalized by endocytic trafficking. Intracellular MUC1-C homodimers are transported to the mitochondrial outer membrane, where they block cell death. MUC1-C homodimers are also imported to the nucleus by importin-β and interactions with the nuclear pore complex (NPC). In the nucleus, MUC1-C interacts with TFs, such as WNT/β-catenin/TCF4, NF-κB, NOTCH and MYC, to drive EMT, epigenetic reprogramming and the cancer stem cell (CSC) state, which contribute to lineage plasticity, DNA damage resistance and immune evasion. Antibodies generated against the MUC1-C extracellular domain are being developed for CAR T cell and antibody–drug conjugate (ADC) immunotherapeutics. Targeting the MUC1-C cytoplasmic domain with the GO-203 inhibitor blocks MUC1-C homodimerization, nuclear import and function. Figure modified from [57,58].
Figure 1. MUC1-C integrates activation of TNBC cell membrane signaling with the induction of EMT, lineage plasticity, DNA damage resistance and immune evasion. Activation of MUC1-C by loss of homeostasis is associated with the formation of homodimers that interact with EGFR and other RTKs at the cell membrane. MUC1-C homodimers are internalized by endocytic trafficking. Intracellular MUC1-C homodimers are transported to the mitochondrial outer membrane, where they block cell death. MUC1-C homodimers are also imported to the nucleus by importin-β and interactions with the nuclear pore complex (NPC). In the nucleus, MUC1-C interacts with TFs, such as WNT/β-catenin/TCF4, NF-κB, NOTCH and MYC, to drive EMT, epigenetic reprogramming and the cancer stem cell (CSC) state, which contribute to lineage plasticity, DNA damage resistance and immune evasion. Antibodies generated against the MUC1-C extracellular domain are being developed for CAR T cell and antibody–drug conjugate (ADC) immunotherapeutics. Targeting the MUC1-C cytoplasmic domain with the GO-203 inhibitor blocks MUC1-C homodimerization, nuclear import and function. Figure modified from [57,58].
Ijms 23 08219 g001
Figure 2. Targeting the MUC1-C extracellular domain. MUC1-C consists of a 58 aa extracellular domain (ED), 28 aa transmembrane domain (TM) and a 72 aa cytoplasmic domain (CD). The MUC1-C ED includes α3 and α4 helices that are exposed with MUC1-C activation and are targets for monoclonal antibodies (MAbs). MAb 3D1 has been generated against the α3 helix and is under development as CAR T cell and ADC approaches. Figure modified from [58,120].
Figure 2. Targeting the MUC1-C extracellular domain. MUC1-C consists of a 58 aa extracellular domain (ED), 28 aa transmembrane domain (TM) and a 72 aa cytoplasmic domain (CD). The MUC1-C ED includes α3 and α4 helices that are exposed with MUC1-C activation and are targets for monoclonal antibodies (MAbs). MAb 3D1 has been generated against the α3 helix and is under development as CAR T cell and ADC approaches. Figure modified from [58,120].
Ijms 23 08219 g002
Figure 3. MUC1-C cytoplasmic domain interacts with diverse effectors of the CSC state. The MUC1-C CD (72 aa) is an intrinsically disordered protein that functions as a node in integrating multiple signaling pathways that promote the CSC state. MUC1-CD includes a CQC motif that is (i) a sensor of increases in ROS, (ii) necessary for the formation of MUC1-C homodimers and (iii) a target for the GO-203 inhibitor. The MUC1-CD CQC motif confers binding to MYC. Shown are tyrosine phosphorylation sites that function as motifs for interactions with PI3K (pYHPM) and GRB2 (pYTNP) in activating the AKT and ERK pathways, respectively. Also shown are potential consensus SH2 binding motifs for interactions with SHC (pYPTY) and PLCγ1 (pYVPP). In addition, MUC1-CD interacts directly with proinflammatory TFs, such as NF-κB and STAT3, and the WNT pathway effectors, β-catenin and TCF4, that are associated with driving the CSC state. Highlighted is the SAGNGGSSLS or SER-rich motif (SRM) that functions as a binding region for STAT3, β-catenin and TCF4. Figure modified from [57,98].
Figure 3. MUC1-C cytoplasmic domain interacts with diverse effectors of the CSC state. The MUC1-C CD (72 aa) is an intrinsically disordered protein that functions as a node in integrating multiple signaling pathways that promote the CSC state. MUC1-CD includes a CQC motif that is (i) a sensor of increases in ROS, (ii) necessary for the formation of MUC1-C homodimers and (iii) a target for the GO-203 inhibitor. The MUC1-CD CQC motif confers binding to MYC. Shown are tyrosine phosphorylation sites that function as motifs for interactions with PI3K (pYHPM) and GRB2 (pYTNP) in activating the AKT and ERK pathways, respectively. Also shown are potential consensus SH2 binding motifs for interactions with SHC (pYPTY) and PLCγ1 (pYVPP). In addition, MUC1-CD interacts directly with proinflammatory TFs, such as NF-κB and STAT3, and the WNT pathway effectors, β-catenin and TCF4, that are associated with driving the CSC state. Highlighted is the SAGNGGSSLS or SER-rich motif (SRM) that functions as a binding region for STAT3, β-catenin and TCF4. Figure modified from [57,98].
Ijms 23 08219 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yamashita, N.; Kufe, D. Addiction of Cancer Stem Cells to MUC1-C in Triple-Negative Breast Cancer Progression. Int. J. Mol. Sci. 2022, 23, 8219. https://doi.org/10.3390/ijms23158219

AMA Style

Yamashita N, Kufe D. Addiction of Cancer Stem Cells to MUC1-C in Triple-Negative Breast Cancer Progression. International Journal of Molecular Sciences. 2022; 23(15):8219. https://doi.org/10.3390/ijms23158219

Chicago/Turabian Style

Yamashita, Nami, and Donald Kufe. 2022. "Addiction of Cancer Stem Cells to MUC1-C in Triple-Negative Breast Cancer Progression" International Journal of Molecular Sciences 23, no. 15: 8219. https://doi.org/10.3390/ijms23158219

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop