Next Article in Journal
Characterization and Mapping of a Rolling Leaf Mutant Allele rlT73 on Chromosome 1BL of Wheat
Previous Article in Journal
DKK3 Expression in Glioblastoma: Correlations with Biomolecular Markers
Previous Article in Special Issue
Cancer Stem Cells from Definition to Detection and Targeted Drugs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Informed by Cancer Stem Cells of Solid Tumors: Advances in Treatments Targeting Tumor-Promoting Factors and Pathways

by
Maya R. MacLean
1,
Olivia L. Walker
1,
Raj Pranap Arun
1,
Wasundara Fernando
1,2 and
Paola Marcato
1,3,4,*
1
Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada
2
Department of Biology, Acadia University, Wolfville, NS B4P 2R6, Canada
3
Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
4
Nova Scotia Health Authority, Halifax, NS B3H 4R2, Canada
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(7), 4102; https://doi.org/10.3390/ijms25074102
Submission received: 28 February 2024 / Revised: 30 March 2024 / Accepted: 2 April 2024 / Published: 7 April 2024
(This article belongs to the Special Issue The Hallmarks of Cancer Stem Cells)

Abstract

:
Cancer stem cells (CSCs) represent a subpopulation within tumors that promote cancer progression, metastasis, and recurrence due to their self-renewal capacity and resistance to conventional therapies. CSC-specific markers and signaling pathways highly active in CSCs have emerged as a promising strategy for improving patient outcomes. This review provides a comprehensive overview of the therapeutic targets associated with CSCs of solid tumors across various cancer types, including key molecular markers aldehyde dehydrogenases, CD44, epithelial cellular adhesion molecule, and CD133 and signaling pathways such as Wnt/β-catenin, Notch, and Sonic Hedgehog. We discuss a wide array of therapeutic modalities ranging from targeted antibodies, small molecule inhibitors, and near-infrared photoimmunotherapy to advanced genetic approaches like RNA interference, CRISPR/Cas9 technology, aptamers, antisense oligonucleotides, chimeric antigen receptor (CAR) T cells, CAR natural killer cells, bispecific T cell engagers, immunotoxins, drug-antibody conjugates, therapeutic peptides, and dendritic cell vaccines. This review spans developments from preclinical investigations to ongoing clinical trials, highlighting the innovative targeting strategies that have been informed by CSC-associated pathways and molecules to overcome therapeutic resistance. We aim to provide insights into the potential of these therapies to revolutionize cancer treatment, underscoring the critical need for a multi-faceted approach in the battle against cancer. This comprehensive analysis demonstrates how advances made in the CSC field have informed significant developments in novel targeted therapeutic approaches, with the ultimate goal of achieving more effective and durable responses in cancer patients.

1. Introduction

In the last twenty years, cancer recurrence, metastasis, and chemoresistance have increasingly been associated with cancer stem cells (CSC). CSCs are a subpopulation of cancer cells defined by their proclivity for self-renewal and differentiation, as well as tumorigenicity [1,2,3]. Indeed, an isolated population of CSCs could give rise to new tumors, while simultaneously repopulating the tumor with both CSC and non-CSC populations [1]. CSC populations can be defined by different cell surface and intracellular markers [4]. Interestingly, despite their role as aggressive tumor initiators, CSCs only comprise a small percentage of bulk tumor volume, with reports suggesting ranges from 0.02% to 25% [5,6]. The importance of CSCs in the development of chemoresistance has been demonstrated in cancers including breast, head and neck, mesothelioma, gastric, and ovarian cancers and have been extensively reviewed elsewhere [7,8,9,10,11]. Furthermore, CSCs have also been implicated as important contributors to metastasis across multiple cancer types [12,13,14,15,16,17,18]. Many current therapies that are designed to target fast proliferating bulk tumor cells are much less effective at clearing the highly aggressive and possibly quiescent CSC populations, which can ultimately lead to metastasis and recurrence [19,20,21]. Thus, developing therapies that can target aggressive CSC populations has been of increasing interest in the last 20 years.
There are several CSC markers and associated pathways thar are involved in maintaining stemness and the aggressive CSC phenotype across multiple cancers and are potential candidates as anti-cancer targets. In this review, we discuss the status of the therapeutics available that target CSC markers and pathways more active in CSCs across multiple cancer types, focusing on solid tumors. We include discussion of advances in targeting aldehyde dehydrogenases (ALDHs), epithelial cell adhesion molecule (EpCAM), Cluster differentiation 44 (CD44), CD55, C-X-C receptor 4 (CXCR4), CD133, Nanog, neurogenic locus notch homolog protein (Notch), Wnt/β-catenin, Sonic hedgehog (SHH), and Sry-box 2 (SOX2). Figure 1 represents the cellular location of the CSC markers and factors/pathways associated with CSCs that are discussed in this review.
It is notable that not all the molecules included in this review are necessarily CSC markers (e.g., CXCR4), but we have included them because they are enriched in CSCs or promote CSC aggressiveness/maintenance, and as such, targeting them could reduce CSCs. Furthermore, the properties of tumor progression and chemoresistance that have been ascribed to these molecules are also intrinsically associated with CSCs and cannot be separated from CSCs. In the reviewed studies, drugs that block tumor progression and chemoresistance are also described to inhibit CSC populations. Furthermore, in our review of drug tumor studies, not all studies specifically addressed effects on the CSC population; however, for completeness, we included those studies. In general, the molecules that are included in this review are drug targets outside of their association with CSCs because they promote tumor progression and chemoresistance. Finally, we use the term CSC, which is often equated to tumor-initiating cell (TIC); however, TICs are not necessarily CSCs and vice-versa. TIC is a functional term defined by the ability to form new tumors in a suitable host organism. CSCs may not necessarily have this ability. CSCs display stemness properties, which TICs may not have.

2. Aldehyde Dehydrogenases

ALDHs are a group of nicotinamide adenine dinucleotide (NAD+) or NAD phosphate (NAD+) dependent enzymes that catalyze the irreversible oxidation of aldehydes to carboxylic acids. The ALDH superfamily is composed of 19 different isoforms, which exhibit distinct tissue expression profiles and subcellular location, as well as substrate specificity and function. In general, ALDHs serve as important regulators of cellular homeostasis, removing toxic aldehydes that are produced during metabolic processes, such as those arising from amino acid catabolism, lipid peroxidation, and exogenous xenobiotics [22]. Additionally, there are several isoforms (i.e., ALDH1A1, ALDH1A2, ALDH1A3) that are responsible for converting the vitamin A metabolite, retinaldehyde, to retinoic acid (RA), and have been associated with CSCs [23,24]. Retinoic acid can modulate the expression of genes, including those responsible for apoptosis, cellular proliferation, and differentiation [25,26].
Due to their role in supporting cellular homeostasis and metabolism, it is unsurprising that there are several ALDH isoforms that have been implicated in cancer progression as CSC markers. ALDH activity in cancer stem cells is most commonly detected by the Aldefluor assay, which measures the conversion of BODIPY aminoacetaldehyde to fluorescent reaction product BODIPY aminoacetate [23]. High ALDH activity can be attributed to several ALDH isoforms, including ALDH1A1, ALDH1A2, ALDH1A3, ALDH2, and ALDH3A1 [27,28,29,30]. It is well-appreciated that these isoforms are involved in maintaining the stemness of CSCs across multiple cancer types, including breast, prostate, colorectal, pancreatic, melanoma, lung, liver, and brain [30,31,32,33,34,35,36]. Indeed, ALDH isoforms have been linked to enhanced clonogenicity and tumorigenicity, as well as chemoresistance and metastasis [13,30,31,33,37]. Recently, there has been increasing interest in the development of ALDH inhibitors to treat cancer and target aggressive CSC populations. The following subsections discuss each of the ALDH isoforms that have been linked to CSCs followed by the development of inhibitors that target these isoforms specifically. We also include a subsection on inhibitors that have pan-ALDH inhibitory activity. Table 1 provides a summary of the inhibitors.

2.1. ALDH1A1

ALDH1A1 has been linked to stemness features such as chemotherapy resistance and worse prognosis in glioma, prostate, esophageal, gastric, ovarian, and breast cancer [30,35,38,39,40,41]. Several ALDH1A1-specific inhibitors have been developed, including NCT-501, CM37, and 974. In head and neck squamous cell carcinoma (HNSCC), ALDH1A1 was enriched in cisplatin and 5-fluorouacil-resistant squamous cell carcinoma (SCC) Cal-27 cell lines, which displayed enhanced stemness features such as spheroid formation, clonogenicity, and migratory potential [42]. Treatment with 40 nM and 80 nM of NCT-501, a theophylline-based reversible inhibitor of ALDH1A1, significantly reduced Cal-27 spheroid size and formation, as well as migratory capacity [42,43]. Furthermore, intra-tumoral injection of 100 μg of NCT-501 in Cal-27 cisplatin-resistant xenografts in mice inhibited tumor growth by 78% compared to control and induced sensitivity to cisplatin in patient-derived explant samples [42]. However, NCT-501 exhibited low cellular activity in pancreatic and colorectal cancer cell lines, MIA PaCa-2 and HT-29, with an IC50 > 4 μM, indicating that its effect may be cell-line specific [44]. CM37, another small molecule ALDH1A1 inhibitor, reduced spheroid formation in ovarian cancer cell lines OVCAR8 and OVCAR3 at 5 and 20 μM, respectively; however, an IC50 has yet to be determined [45]. In another recent study, Compound 974 demonstrated an IC50 of 470 nM for binding with purified ALDH1A1 [46]. Pre-treatment of OVCAR3 cells with 5 μM of Compound 974 inhibited tumor formation in vivo in a limiting dilution analysis, indicating a reduction in CSC frequency [46]. Furthermore, Compound 974 inhibited chemotherapy-induced senescence in OVCAR3 cells [46].

2.2. ALDH1A3

It is well appreciated that ALDH1A3 is an important driver of stemness in multiple cancer types, including breast, melanoma, glioblastoma, prostate, non-small cell lung cancer (NSCLC), head and neck cancer, and colon cancer [47]. Indeed, ALDH1A3 has been shown to enhance metastasis, clonogenicity, and chemoresistance [48,49]. There are a few compounds that show specificity toward ALDH1A3, including MF-7, GA11, MCI-INI-3, NR6, and YD1701 [12,50,51,52,53]. Intraperitoneal injections of GA11 (20 mg/kg) attenuated the growth of mesenchymal glioma tumor-derived MES83 and MES267 cells in mice [50]. Later, modifications of GA11 to MF-7 increased its IC50 in cell-free assays from 4.7 μM to 22.8 μM; however, MF-7 displayed increased functional efficacy compared to GA11 when tested for anti-proliferative activity in the triple-negative breast cancer (TNBC) cell line MDA-MB-468 [12]. While intraperitoneal injections of MF-7 reduced formation and size of MDA-MB-231 and MDA-MB-468 brain metastases, there was a paradoxical increase in the number of lung and bone metastases [12]. NR6, an imidazo [1-2-a] pyridine derivative, reduced cell growth and the invasiveness of U87MG glioblastoma and HCT116 colorectal cancer (CRC) cell lines [51]. Recently there have been improvements in the design of other ALDH1A3-specific inhibitors that are able to prevent pocket binding of the ALDH1A3 substrate, retinal. MCI-INI-3 is one such compound designed in silico that reduces RA production in the glioma stem cell line, U87MG, at 15 μM [52]. However, MCI-INI-3 displays cytotoxic effects at 15 μM on non-ALDH1A3-expressing glioma stem cell lines, indicating that it may not have an ALDH1A3-specific impact on glioma stem cell viability [52]. In silico analyses have also revealed YD1701 as a novel ALDH1A3 inhibitor with a reported IC50 of 12.0824 μg/mL [53]. Indeed, YD1701 reduced the invasive capacity of colorectal cancer cell lines SW480, HT29, HCT116, and CRC1 at 0.04 μg/mL and prolonged survival in HCT116-tumor-bearing mice [53].

2.3. ALDH2

ALDH2 has been identified as a CSC marker in liver and lung cancers [34,54]. Compared to the other ALDH isoforms, relatively few reports have been published on ALDH2 inhibitors, and fewer still have been tested as anti-cancer agents. CVT-10216 is a selective, reversible inhibitor of ALDH2 (IC50 = 0.029 μM), which was initially developed as a method for treating alcoholism [55]. Similarly, daidzin, an isoflavone derived from the kudzu vine, has an IC50 of 80 nM and has been shown to reduce alcohol consumption in animal models [56,57]. In transwell migration assays, 25 μM of CVT-10216 reduced the migratory capacity of colorectal cancer cell lines HCT-116 and DLD-1 cell lines [13]. Similarly, 100 μM or 25 μM of daidzin reduced DLD-1 and HCT116 migration in transwell migration assays, respectively [13]. Furthermore, both inhibitors reduced the clonogenicity of the HCT-116 and DLD-1 cell lines; however, CVT-10216 exhibited increased efficacy as it reduced clonogenicity at 25 μM in HCT-116 cells, compared to 100 μM with daidzin [13].

2.4. ALDH3A1

It is well appreciated that ALDH3A1 is linked to chemotherapy resistance, particularly for agents such as cyclophosphamide, paclitaxel, and doxorubicin [58,59]. ALDH3A1 inhibitors such as dyclonine, CB7, and CB29 have all been shown to enhance cancer cell sensitivity to sulfasalazine and cyclophosphamide, respectively [60,61,62]. Indeed, 50 μM of dyclonine, an oral anesthetic, was shown to inhibit ALDH activity in HSC-4 cells and enhanced sulfasalazine cytotoxicity in ALDH3A1+ gastric tumor cells isolated from K19-Wnt1/C2mE mice [60]. Others have reported an IC50 value of 76 μM for dyclonine against ALDH3A1 activity, and while dyclonine has also been reported to inhibit ALDH2, sulfasalazine resistance is attributed to ALDH3A1 expression, not ALDH2 [60,63]. CB57, another ALDH3A1 inhibitor, was shown to be highly specific for ALDH3A1, with an IC50 of 0.2 μM, and did not display any inhibitory effects on any of the ALDH1 or ALDH2 isoforms [61]. Indeed, CB57 sensitized lung and glioblastoma cell lines, A549 and SF767, to mafosamide treatment, evidenced by a significant reduction in cell proliferation when compared to treatment with mafosamide alone [61]. Similarly, CB29, another small molecule inhibitor of ALDH3A1, displayed an IC50 of 16 μM, and decreased the ED50 of mafosamide 1.6-fold in SF767 cells [62]. Using chemoproteomics-enabled covalent ligand screening, Counihan et al. identified EN40, a selective ALDH3A1 inhibitor with an IC50 of 2 μM [64]. Indeed, 10 μM of EN40 impaired survival in lung cancer cell line A549 and significantly reduced tumor growth in A549 at a daily dose of 50 mg/kg [64].

2.5. Pan-ALDH Inhibitors

As the 19 ALDH isoforms share up to 40% sequence homology, there are several non-specific ALDH or pan-ALDH inhibitors [22,65]. N,N diethylaminobenzaldehyde (DEAB) is a well-known ALDH inhibitor and has been shown to inhibit ALDH1A1 (IC50 = 0.057 μM), ALDH1A2 (IC50 = 1.2 μM), ALDH1A3 (IC50 = 3 μM), ALDH1B1 (IC50 = 1.2 μM), ALDH2 (IC50 = 0.16 μM), and ALDH5A1 (IC50 = 13 μM) [66]. In melanoma, 200 mg/kg of DEAB has been shown to reduce melanoma xenograft growth and the number of residual melanoma cells [67]. Similarly, DEAB inhibited colony-forming ability in pancreatic cancer cell lines BxPC3 and Panc1 [68]. An in silico screen identified another multi-isoform inhibitor, KS100, which inhibits ALDH1A1 (IC50 = 207 nmol/L), ALDH2 (IC50 = 1410 nmol/L), and ALDH3A1 (IC50 = 240 nmol/L) [69]. Nanoliposomal encapsulation of KS100 (hereafter referred to as NanoKS100) inhibited UACC 903 and 1205-Lu derived melanoma xenograft tumor growth at a dose of 20 mg/kg [69]. Dimethyl ampal thiolester (DIMATE) is an irreversible inhibitor of ALDHs 1 and 3 [70]. DIMATE exhibited cytotoxic effects on acute myeloid leukemia (AML) cell lines, with IC50s ranging between 1.67 μmol/L to 12.2 μmol/L and, interestingly, had no cytotoxic effects on healthy hematopoietic stem cells [71]. Furthermore, 14 and 28 mg/kg of DIMATE significantly reduced the numbers of circulating xenografted AML cells in immunodeficient mice [71]. Another pan-ALDH1 inhibitor, 637A, inhibits ALDH1A1 (IC50 = 246 nM), ALDH1A2 (230 nM), and ALDH1A3 (348 nM) induced calcium-dependent necroptosis in ovarian cancer cell line A2780 [72]. Interestingly, 637A also improves response to cisplatin in SKOV3 cells [72]. Citral has also been shown to inhibit several ALDH isoforms, including ALDH1A3 and ALDH2 [73]. In SKBR3 cells, 10 μM of citral significantly reduced ALDH2-mediated Aldefluor fluorescence, while only 1 μM was required to reduce ALDH1A3-mediated Aldefluor activity in MDA-MB-231 cells [73]. Furthermore, 0.4 mg/kg of nanoparticle encapsulated citral reduced MDA-MB-231-derived tumor growth in mice [73]. Interestingly, disulfiram, a well-known treatment for alcohol abuse, has recently been suggested as a repurposed anti-cancer drug due to its inhibition of ALDH isoforms such as ALDH1A1, ALDH1A3, and ALDH2 [73,74,75]. Indeed, 10 μM of disulfiram induces apoptosis of TNBC MDA-MB-231 ALDH1A3 overexpressing cells [73]. Similarly, a complex of disulfiram with copper reduced ALDH2 expression in lung cancer A549 cells and enhanced chemosensitivity to taxol [75].
The intricate involvement of ALDHs in cellular homeostasis and metabolism highlights their significance in normal and physiological processes. However, the role of specific ALDH isoforms, such as ALDH1A1, ALDH1A3, ALDH2, and ALDH3A1, as CSC markers has shifted attention towards developing targeted inhibitors to prevent cancer progression. While there is an abundance of evidence highlighting the use of both pan and isoform-specific ALDH inhibitors as anti-cancer agents, none have progressed into clinics, namely, due to working concentrations that surpass those required for clinical translation. The versatility of ALDH inhibitors in targeting multiple cancer types emphasizes their potential as a valuable addition to the armamentarium of cancer therapy, likely as adjuvant agents. The ongoing preclinical research and the development of highly specific and potent ALDH inhibitors will be pivotal in assessing their translational potential.

3. EpCAM

Epithelial cell adhesion molecule (EpCAM) is a type I transmembrane protein, and in the early research, it was classified as a cell adhesion molecule (CAM) due to its role in mediating cell–cell adhesion [76]. While most CAMs display broad tissue distribution, EpCAM is restricted to epithelial tissue in healthy individuals and is generally located on the basolateral or basal cell membrane [77,78]. The role of EpCAM in cancer was first elucidated in 1979, when it was described as a tumor antigen in colorectal carcinomas [79]. Indeed, EpCAM is highly expressed in lung, colon, intestine, breast, and prostate carcinoma [80]. Beyond its proposed role as a CAM, EpCAM has also been implicated as a CSC marker and mediator of cell proliferation, migration, and gene expression in cancer [80,81,82,83]. In breast cancer, EpCAM-positive cells display CSC characteristics, with potential for self-renewal and differentiation, as well an enhanced tumorigenicity and migration [82]. Similarly, isolated EpCAM+ HCC cells initiated highly invasive HCC in mice and exhibited similar cancer-stem-cell-like traits [84]. In AML, EpCAM+ K562 and HL60 cells are resistant to chemotherapy and display enhanced tumorigenicity, compared to EpCAMlow cells [85]. Furthermore, EpCAM+ ovarian cancer cells exhibited CSC characteristics of tumorigenicity and migration and were resistant to platinum-based chemotherapy [86,87]. However, increased EpCAM expression has also been linked to improved patient survival in some cancers, including renal clear cell carcinoma and thyroid carcinoma, indicating its role as a cancer promoter or suppressor may be cancer-type specific [88,89]. Regardless, the abundance of evidence implicating EpCAM as a CSC marker and its role in promoting aggressive cancer implicate it as an attractive target for anti-cancer agents. Several agents have been developed, including small molecule inhibitors, immunotherapy agents, monoclonal antibodies (mAbs), and aptamers. Table 1 provides a summary of the inhibitors.

3.1. Antibodies

As EpCAM is primarily expressed on the surface of cancer cells, there has been great interest in the development of EpCAM mAbs. EpAb2-6, a highly specific EpCAM antibody, induced apoptosis via inhibition of EpCAM signaling in HCT116, SAS, AsPC-1, and SW620 cells, an effect that was diminished in HCT116/shEpCAM and normal cells [90]. Co-treatment of EpAb2-6 (20 mg/kg) alone and in combination with IFL (irinotecan, leucovorin, and fluorouracil (5FU)) in mice bearing colon cancer xenografts significantly decreased tumor volume and improved survival times [90]. In a similar study, EpAb2-6 (20 mg/kg) in combination with atezolizumab, an anti-programmed cell death ligand 1 (PD-L1) antibody, nearly eradicated tumors in peripheral blood mononuclear cell (PBMC)-H441-xenografted mice and increased the CD8+ T cell infiltration compared to mice treated with atezolizumab or EpAb2-6 alone [91]. Adecatumumab (MT201) is a human anti-EpCAM mAb that induces cell death via antibody-dependent cellular toxicity and displays intermediate affinity for EpCAM, which is generally thought to be more well-tolerated than high-affinity antibodies [92]. EpCAM+ ovarian cancer cell lines established from patients with chemotherapy-resistant disease (OSPC-ARK-1, OSPC-ARK-2, OSPC-ARK-4, CC-ARK1, and CC-ARK-2) treated with 5 μg/mL of adecatumumab displayed increased cytotoxicity compared to control [92]. Adecatumumab has also been tested in several Phase I and II clinical trials and is generally well-tolerated [93,94,95]. In breast cancer patients, adecatumumab reduced the incidence of metastasis in patients with high EpCAM expression but did not result in partial or complete response in any of the patients [93]. Similarly, adecatumumab delayed disease progression in prostate cancer patients with EpCAM+ tumors and rising prostate-specific antigen (PSA) levels after prostatectomy [94]. A novel anti-EpCAM antibody, AM-928, is currently recruiting for the first-in-human Phase-I study (NCT05687682) [96]. Recently, there has been increasing interest in the use of bispecific T-cell-engaging (BiTE) antibodies to facilitate T-cell recruitment and activation [97]. Solitomab (MT110) is a BiTE antibody construct that binds to EpCAM and CD3 [98]. Indeed, pancreatic cancer cells AL6 and 185, co-cultured with (PBMCs) and 100 ng/mL of solitomab, significantly reduced spheroid formation compared to PBMCs alone [98]. Furthermore, in vivo treatment of solitomab in mice with PBMC co-cultured A6L-derived tumors resulted in complete abrogation in tumor forming ability, indicating that the tumorigenic CSC population had been effectively eradicated [98]. Ovarian cancer cell lines with high EpCAM expression have been shown to be resistant to T-cell-mediated killing; however, following incubation with solitomab (1 μg/mL), they became highly sensitive to T-cell-mediated killing [99]. Despite promising results in vitro and in murine models, solitomab has displayed relatively modest results in humans. Indeed, in a phase I trial for solitomab on refractory solid tumors, the best outcome among patients was confirmed stable disease (NCT00635596) [100]. Catumaxomab is a similar BiTE antibody construct targeting EpCAM and CD3 that was initially approved in the European Union in 2009 for intraperitoneal treatment of malignant ascites but was withdrawn for commercial reasons in 2017 [101]. Interestingly, a recent paper on mRNA-lipid nanoparticle (LNP) delivery of EpCAM-CD3 BiTEs shows promise in vivo [102]. Intratumoral injection of EpCAM-CD3 hFc mRNA-LNPs (1 μg/mouse) in combination with intravenous injection of T cells significantly decreased OVCAR-5 tumor growth in mice [102].

3.2. CAR-T Therapies

Chimeric antigen receptor redirected T cells (CAR-T) are another promising avenue of targeting EpCAM in cancer. In EpCAM+ ovarian cancer cell line SW626, anti-EpCAM DNA CAR-T cells displayed high lysis activity, being able to kill 91.5% of SW626 cells [103]. Furthermore, just one intraperitoneal injection of anti-EpCAM DNA CAR-T cells in mice bearing SKOV3-Luc ovarian tumors resulted in decreased tumor burden and increased survival [103]. Adoptive transfer of peripheral blood lymphocytes (PBL) transduced with an EpCAM-specific CAR prevented metastasis of prostate cancer PC3 cells and significantly reduced tumor growth of PC3M cells in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice [104]. A recent publication examined the ability of anti-EpCAM CAR-T cells to prevent lung cancer brain metastasis [105]. While intraparenchymal injection of anti-EpCAM CAR-T cells reduced LL/2 tumor growth in the brain in vivo, intravenous administration of the CAR-T cells failed to accumulate within the tumor and, in both cases, failed to persist within the tumor, suggesting that the success of CAR-T cells may be cancer- and location-specific [105]. Anti-EpCAM CAR-T cells have also been shown to produce robust anti-tumor responses in gastric cancer SNU-638 and MKN-45 and pancreatic Capan2-Fluc+ models in mice, with complete responses maintained in nearly all the mice [106]. Despite the relative success of CAR-T cells as anti-cancer agents, cancer relapse may still occur and often manifests in antigen escape [107]. Thus, a novel mechanism to circumvent antigen escape is through bispecific CAR-T cells, which target two antigens. Indeed, bispecific EpCAM and intercellular adhesion molecule-1 (ICAM-1) CAR-T cells completely cleared subcutaneous SNU-638 tumors in mice and prevented relapse more durably than anti-EpCAM CAR-T cells alone [106]. Several clinical trials have begun due to the success of anti-EpCAM CAR-T cells in vitro and in mouse models. IMC001, an anti-EpCAM CAR-T therapy, as monotherapy or in combination with immune checkpoint inhibitors, is currently in Phase I and II trials for EpCAM+ tumors of the digestive system (NCT05028933, NCT04196465). The preliminary results released in early 2023 for the phase I trial indicated that of the six gastric cancer patients, two achieved partial response, while three were evaluated as having stable disease [108].

3.3. Immunotoxins

In a similar vein, the use of immunotoxins to target EpCAM has also been of interest. VB6-845 is an anti-EpCAM fragment antigen-binding (Fab) region linked to DeBouganin, a plant-derived toxin that inhibits protein translation through inactivating RNA N-glycosidase activity [109]. In vitro, VB6-845 displayed high cytotoxicity against EpCAM+ cancer cell lines NIH:OVCAR-3, Caov-3, MCF-7, NCI-H69, HT29, and CAL 27 with nanomolar IC50 values (0.4–1.8 nM). In vivo experiments with NIH:OVCAR-3 tumor bearing mice revealed VB6-845 (10 and 20 mg/kg) has potent anti-tumor activity, significantly decreasing tumor volume and resulting in 100% survival in NOD/SCID mice [110]. In phase I clinical trials (NCT00481936), VB6-485 showed modest efficacy, with decreases in tumors ranging from 4 to 15% in a patient with breast carcinoma and 11 to 29% in a patient with renal cell carcinoma [110,111]. A similar recombinant immunotoxin, VB4-845, is an anti-EpCAM scFv fragment linked to Pseudomonas exotoxin A (ETA) [112]. Intratumoral injections of VB4-845 in combination with Nivolumab reduced tumor growth in mice, likely via the complimentary action of VB4-845-mediated immunogenic cell death (ICD) [113]. In a phase I trial of VB4-845 in patients with recurrent, advanced HNSCC, there were several instances of partial response and at least one patient with confirmed complete response [114]. As of 2008, VB4-845 was reportedly undergoing phase II clinical trials in HNSCC, as well as phase III trials for the treatment of superficial transitional cell carcinoma of the bladder (NCT04859751) [115,116]. A more recently developed anti-EpCAM immunotoxin, scFv2A9-PE, similarly uses the Pseudomonas aeruginosa exotoxin and exhibits an IC50 of 50 pM for cytotoxicity in EpCAM+ HHCC hematopoietic cells; however, it has yet to be tested against cancer cells [117].

3.4. Pharmacologic Inhibitors

As evidenced, immunomodulatory agents have been of great interest in the targeting of EpCAM to treat cancer. However, relatively few pharmacological inhibitors against EpCAM have been developed. Recently, high-content screening of a small molecule compound library yielded several robust inhibitors of EpCAM signaling [118]. Indeed, several of the compounds reduced EpCAM-mediated cyclin D1 (CCND1) expression in colorectal carcinoma cell line HCT-8; however, they have yet to be tested against tumor growth [118]. Other mechanisms of pharmacological inhibition of EpCAM have targeted EpCAM-related pathways instead. SyntOFF, an inhibitor of the syntenin PDZ2 domain, significantly decreased the loading of EpCAM into exosomes and reduced mammosphere formation in MCF-7 cells at 50–100 μM [119].

3.5. Antibody-Drug Conjugates

Interestingly, other groups have used anti-EpCAM antibody–drug conjugates (ADCs) to target cancer cells. The novel therapeutic ADC α-amanitin-glutarate-chiHEA125 (chiHEA125-Ama) is an anti-EpCAM mAb conjugated to the mushroom toxin α-amanitin, which inhibits DNA transcription [120]. Administration of a single intraperitoneal injection of chiHEA125-Ama (50 μg/kg) dramatically decreased pancreatic BxPc-3 tumor volume in mice, resulting in complete tumor regression in 50% of the mice [120]. While tumor relapse was observed in 80% of the mice receiving 50 μg/kg of chiHEA125-Ama, 100 μg/kg of chiHEA125-Ama prevented tumor relapse in close to 40% of the mice tested, suggesting the eradication of tumor initiating cells [120].

3.6. Other EpCAM-Targeting Strategies

Recently, there has been increasing interest in the use of aptamers and RNA interference (RNAi) to target cancer. Aptamers are single stranded RNA or DNA oligonucleotides that can bind to targets including proteins, nucleic acids, and organic molecules [121]. Anti-EpCAM aptamers can be used to deliver therapeutics to EpCAM+ cells. Interestingly, a comparison between anti-EpCAM antibodies versus anti-EpCAM aptamers revealed that aptamers display better tumor penetration and retention in HT29 tumors in mice [122] Indeed, in a 2016 study, an anti-EpCAM aptamer was used to deliver small interfering RNA (siRNA), which reduced EpCAM mRNA expression and protein levels, as well as reducing viability in breast MCF-7 and retinoblastoma WERI-Rb1 cells [123]. Alternative mechanisms of siRNA delivery include the conjugation of anti-EpCAM mAbs to polyetheleneimine (PEI)-capped gold nanoparticles (AuNP) loaded with EpCAM siRNA [124]. In this study, internalization of the EpCAM antibody-conjugated AuNP-PEI and delivery of the EpCAM siRNA resulted in a significant downregulation of EpCAM gene expression and viability in retinoblastoma Y79 cells [124].
Thus, it is evident that the role of EpCAM in cancer biology extends beyond its initial characterization as a CAM to encompass crucial functions as a CSC marker. The development of diverse agents ranging from mAbs and immunotoxins to CAR-T cells and pharmacological inhibitors reflects the concerted efforts to exploit EpCAM’s significance as a contributor to stemness across multiple cancers. Despite these advancements, the complexity of EpCAM necessitates continued exploration and refinement of EpCAM-targeted therapies for translation into the clinical setting and improving outcomes for patients with EpCAM-expressing malignancies.

4. CD44

CD44 is a transmembrane glycoprotein that displays broad tissue distribution and expression. CD44 can bind to several ligands to influence numerous cellular processes, including hyaluronic acid (HA), fibronectin, osteopontin (OPN), collagen, and matrix metalloproteinase-9 (MMP-9) [125,126]. CD44-HA binding induces conformational changes in the CD44 structure, allowing binding of cytoskeletal elements and adaptor proteins, which can lead to activation of signaling pathways involved in proliferation, migration, invasion, and adhesion [125,127]. It is well-appreciated that CD44 is an important prognostic marker for CSCs and is one of the most widely recognizable CSC markers across cancer types. Indeed, CD44 is a marker for CSCs in lung, breast, gastric, liver, CRC and AML cancers [128,129]. CD44 expression has been linked to the epithelial–mesenchymal transition (EMT), which is an important feature of stemness in CSCs [130]. Indeed, CD44+ pancreatic ductal adenocarcinoma (PDAC) cells with an EMT phenotype display increased tumorigenicity and resistance to gemcitabine compared to CD44 cells [131]. In 2003, Al-Hajj et al. identified a CD44+CD24−/lowLineage of breast cancer cells isolated from breast cancer patients [1]. This CD44+ subpopulation displayed incredible tumorigenicity, being able to form tumors in mice with as few as 100 cells [1]. Furthermore, serial passaging of the isolated CD44+ population gave rise to not only the highly tumorigenic CD44+ cells but also a mixed population of cells that form the bulk of tumors, highlighting the role of CD44 in maintaining features of stemness in CSCs [1]. In gastric cancer cells, short hairpin RNA (shRNA) knockdown of CD44 reduced spheroid formation in vitro, as well as tumorigenicity in vivo [132]. Furthermore, CD44+ gastric cancer cells also displayed enhanced resistance to chemotherapy and radiation-induced cell death [132]. Thus, developing therapeutics to target CD44 is of great interest in the pursuit to eliminate CSC populations and improve survival in patients (summarized in Table 1).

4.1. Antibodies

Several mAbs against CD44 have been developed as cancer therapeutics. Bivatuzumab (BIWA-4) is an anti-CD44 mAb conjugated to microtubule inhibitor mertansine that was tested in HNSCC patients (NCT02254018) [133]. While three patients experienced partial response, significant skin toxicities with a fatal outcome occurred in one trial, and clinical trials were halted [133]. Pre-treatment of another mAb, H4C4, on pancreatic PANC-1 cells significantly decreased tumor formation and reduced clonogenicity in vitro [134]. Furthermore, in a post-radiation tumor recurrence model, only 25% of mice exhibited recurrent tumor growth when treated with H4C4, compared to 90% of mice treated with radiation alone, indicating that the CD44+ tumor initiating population had been eradicated by H4C4 treatment [134]. Interestingly, H4C4 also downregulated the expression of several CSC genes, including Sox2 and Nanog [134]. In MCF-7 cells with acquired invasive potential, CD44 is significantly upregulated [135]. However, treatment with IM7, a CD44 mAb, reduced the invasive and migratory capacity of aggressive MCF-7 cells and MDA-MB-231 cells [135]. In a chronic lymphocytic leukemia model, human leukemia stem cells (LSC) from imatinib-responsive or -resistant patients were transplanted into immunodeficient mice [136]. Treatment with RG7356 (RO5429083), a humanized CD44 mAb, reduced the number of LSCs in bone marrow in both the imatinib-responsive and -resistant mice and sensitized the LSCs to dasatinib, a kinase inhibitor [136]. Interestingly, RG7356 was also shown to induce internalization of cell-surface CD44 in chronic lymphocytic leukemia (CLL) cells, and treatment with 0.01 mg/kg of RG7356 in CLL xenografted mice resulted in complete clearance of the leukemia cells [137]. Furthermore, RG7356 displayed remarkable tumor inhibition in HNSCC CAL 27 xenografts in mice [138]. Interestingly, RG7356 also expanded the NK cell population in PMBCs derived from healthy donors [138]. RG7356 has also been tested in phase I clinical trials for patients with solid tumors (NCT01358903) and AML (NCT01641250). While RG7356 was generally well-tolerated in both studies, the majority of patients experienced cancer progression. In the solid tumor clinical trial, the best response recorded was stable disease, which accounted for 21% of patients enrolled [139]. Conversely, in patients with AML, at least one complete response and partial response were recorded [140].

4.2. NIR-PIT

In recent years, anti-cancer therapy has expanded to include near-infrared photoimmunotherapy (NIR-PIT). NIR-PIT induces targeted, selective death in cancer cells through conjugation of a mAb to a photo-absorber, silica-phthalocyanine (IRDye700DX: IR700) dye [141]. Injection of a CD44 mAb-IR700 dye conjugate (anti-CD44-IR700) in mice bearing oral SCC CD44+ MOC1 and MOC2 tumors significantly inhibited tumor growth and prolonged survival [142]. Interestingly, a similar study combined anti-CD44-IR700 with a systemic anti-PD-1 mAb, resulting in nearly complete eradication of MC38-luc tumors in 70% of the mice [143]. Moreover, this combination treatment also resulted in control of several distant MC38 tumors, indicating that local treatment of one tumor is sufficient to induce systemic anti-cancer effects [143]. Other studies have combined CD44- and CD25-targeted NIR-PIT to simultaneously deplete FOXP3+CD25+CD4+ Treg and CD44+ CSCs in several syngeneic tumor models [144]. Indeed, in MC38-luc-tumor-bearing mice, combined CD44- and CD25-targeted NIR-PIT significantly reduced tumor size and regrowth following treatment [144]. Significantly, in a LL/2 tumor model, just one round of CD44- and CD25-targeted NIR-PIT achieved complete remission in 33% of the mice tested [144]. While clinical trials have begun using cetuximab-IR7000 conjugates (NCT02422979), none have commenced using anti-CD44-IR700 conjugates.

4.3. Other CD44-Targeting Strategies

Other interesting mechanisms of targeting CD44 in cancer include recombinant human proteoglycan 4 (rhPRG4). rhPRG4 has shown success in preserving joint health in vivo in osteoarthritis models, and a recent study demonstrated that it can also suppress transforming growth factor beta (TGFβ)-induced migration and invasion in breast cancer cells by reducing CD44 protein levels [145,146]. It is well-appreciated that TGFβ can increase CD44 expression in breast cancer cells through EMT [147]. Indeed, the TGFβ-induced invasive capacity of MDA-MB-231 cells is at least in part mediated by the HA-CD44 signaling axis [146]. Interestingly, rhPRG4 reduced the TGFβ-induced increase in the CD44 protein level, thus, reducing the invasiveness of breast cancer HCC38 cells [146]. Aptamers and antisense oligonucleotides (ASOs) are another avenue of CD44-targeting that has garnered interest over the past decade. DNA aptamers such as AS1411, which targets nucleolin, have been used in clinical trials with some success [148]. A few DNA aptamers against CD44 have been developed including Apt#7, which inhibited migration of breast HCC38 cells in vitro by preventing interactions between CD44 and erythropoietin-producing hepatocellular receptor tyrosine kinase class A2 (EphA2) [149]. Other approaches have utilized bi-specific aptamers that target CD44 and EpCAM [150]. Intraperitoneal injections (2 nmol/mouse/every two days) of the bi-specific CD44-EpCAM aptamer in mice bearing ovarian OVCAR8 tumors resulted in increased cancer cell apoptosis and a significant reduction in tumor growth [150]. ASOs are single stranded deoxyribonucleotides that bind to mRNA targets and can be used to modify protein expression [151]. The novel CD44 ASO, ASO 4401, sensitized hepatocellular carcinoma SNU-449 cells to doxorubicin and decreased their clonogenicity and invasive capacity [152]. Several phase I and II clinical trials have commenced using ASOs as cancer therapeutics; however, the use of CD44-specific ASOs has not yet been investigated [153].

4.4. Pharmacologic Inhibitors

Several pharmacological inhibitors for CD44 have been developed as anti-cancer agents. The small molecule 1,2,3,4 tetrahydroisoquinoline (THIQ) is a novel inhibitor with affinity for the HA-binding pocket on CD44 [154]. In HNSCC cell lines, treatment with 1 mM of THIQ reduced CD44 protein levels and CD44+ cell population [155]. Interestingly, THIQ treatment also reduced clonogenicity and migratory potential in cisplatin-resistant SCC-131 and CAL-27 cells post cisplatin treatment [155]. Recent modifications of THIQ generated a THIQ-derivative, JE22, which was conjugated to nanoparticles (JE22-NP) [156]. In CD44+ MDA-MB-231 breast cancer cells, JE22-NP displayed a cell viability half maximal efficacy concentration of 49 nM, significantly reducing cell viability [156]. Interestingly, JE22-NP did not exhibit cytotoxic effects on CD44 breast cancer cells or non-cancerous cells. Furthermore, JE22-NP decreased the migratory capacity of MDA-MB-231 cells compared to control [156]. However, whether JE22-NP demonstrates anti-tumor effects in vivo has yet to be investigated. Recently, verbascoside, the active component found in plants from Lamiales order, was identified as a novel CD44 inhibitor which prevents CD44 dimerization and the subsequent release of the CD44 intracellular domain (CD44ICD) [157]. In vitro, 15 μM of verbascoside attenuated the colony formation of glioblastoma U251MG cells and decreased expression of CSC genes Sox2 and Nanog, an effect that was not observed in CD44 PD-GBMC cells [157]. In glioblastoma U251MG models in vivo, verbascoside significantly reduced tumor growth and prolonged survival in mice [157].

4.5. Peptides

Other CD44 inhibitors include A6 (SPL-108), which is a CD44-specific peptide derived from urokinase plasminogen activator (uPA) [158]. While the exact mechanism of A6 has yet to be defined, it is generally thought to bind to CD44 and modulate CD44-mediated signaling [158,159]. The efficacy of A6 as an anti-cancer agent has been tested in several cancers, including prostate, multiple myeloma, breast, and ovarian [159,160,161,162]. Interestingly, A6 appears to be most effective at reducing the migratory capacity of aggressive cancer cell lines. Indeed, A6 blocked migration of the OVCAR3, OVCAR8, IGROV-1, and MDA-MB-468 cell lines, with IC50 values ranging from 15 nmol/L to 65 nmol/L [159]. Interestingly, A6 appeared to potentiate cell binding to HA, a CD44-mediated process, thus halting cell migration [159]. In a metastatic melanoma B16-F10 model, 100 mg/kg of A6 reduced the number of metastatic lesions in the lungs of mice by 50% compared to control [159]. Similarly, in PC-3 LN4 prostate tumor bearing mice, 25% of mice receiving 25 mg/kg of A6 developed metastatic lesions in the lymph nodes, compared to 71% in the control group [160]. Several phase I and II clinical trials for A6 have begun. In a phase II trial for ovarian, fallopian, or peritoneal cancer, none of the thirty-one enrolled patients exhibited response (NCT00939809) [163]. However, in a separate ovarian cancer trial, A6 treatment was associated with delayed clinical progression following first line chemotherapy treatment [164]. The efficacy of A6 has also been tested in phase II trials for chronic lymphocytic leukemia (CLL); however, this trial was terminated due to slow enrollment (NCT02046928). Recently, A6 has been used as a targeting agent to deliver PEGylated liposomal doxorubicin, which decreased C-26 colon cancer tumor volume in mice [165].
The well-established connection between CD44 expression and CSC characteristics, such as tumorigenicity, metastasis, and resistance to therapy, underscores its significance as a prognostic marker and a potential therapeutic target. Encouraging preclinical results, such as the remarkable tumor inhibition by CD44-targeted NIR-PIT and the efficacy of specific inhibitors in reducing metastatic spread, highlight the potential of these approaches. Overall, the multifaceted roles of CD44 in cancer progression and an important regulator of stemness provide a compelling rationale for the continued research and translating these insights into effective therapeutic strategies for cancer management.

5. CD55

CD55 (also known as decay accelerating factor—DAF) is a plasma membrane-bound protein expressed on multiple cell types, including leukocytes, epithelial and endothelial cells, as well as red blood cells. Canonically, CD55 acts as an inhibitor of complement-mediated lysis by degrading the C3 and C5 convertases, which are required for formation of the membrane attack complex [166,167]. In addition, CD55 has a non-canonical function as a CSC-associated factor and marker, promoting cancer progression and chemoresistance in cervical, ovarian, neuroblastoma, breast, and endometrial cancer [166]. Indeed, high CD55 expression in a neuroblastoma is associated with increased sphere-forming efficiency and self-renewal capacity, as well as poor survival outcomes in patients [168]. Similarly, in breast cancer, high CD55 expression in patient tumors is associated with lower relapse-free survival [169]. Furthermore, xenotransplantation of CD55+ MCF-7 breast cancer cells into NOD/SCID mice resulted in significantly increased tumor volume compared to tumors derived from a CD55low population [169]. CD55 is also associated with chemoresistance in endometrioid cancer, as cisplatin resistant CP70 cells displayed high CD55 expression [170]. Interestingly, this CD55+ population also exhibited an enhanced ability for self-renewal, which is a hallmark of stemness [170]. Similarly in cervical cancer, CD55+ C33A cells display significant sphere-forming and migratory abilities, as well as enhanced tumorigenesis and radioresistance [171]. Thus, the exploration of therapeutic strategies targeting CD55 in cancer opens diverse avenues for potential interventions against this critical factor in tumorigenesis and aggressive disease (summarized in Table 1).

5.1. Antibodies

Following the elucidation of CD55 as an important CSC marker, interest in the development of antibodies against CD55 has increased. In 2019, a novel CD55 chimeric mAb displayed efficacy against colorectal cancer cells [172]. This antibody, referred to as anti-CD55, decreased the viability of HT-29 and LoVo cells, likely via activation of the complement system [172]. Furthermore, this anti-CD55 antibody attenuated LoVo tumor growth in mice and decreased in vitro metastatic invasion of DLD-1 cells [172]. In addition, this novel mAb also synergistically enhanced the anti-cancer activity of 5FU in CRC [172]. Another study has reported the use of neutralizing mini-antibodies to CD55 and CD59, MB55 and MB59, respectively, in combination with rituximab, an anti-CD20 antibody in B-cell lymphoma [173]. Interestingly, MB55-MB59 enhanced complement-dependent cytotoxicity and killing of B-cell lymphoma cell lines and significantly improved survival in LCL2 tumor models in mice when used in conjunction with rituximab [173]. In neuroblastoma models, the use of a CD55 neutralizing antibody (CD55 NAb) successfully reduced SKN-BE(2) xenograft tumor growth in mice and improved survival [168]. Interestingly, CD55 NAb also reduced expression of CSC markers NANOG, OCT4, and CD133 in the xenografted SKN-BE(2) tumors, indicating that CD55 targeting may also suppress stemness through multiple mechanisms [168]. In recent years, the use of radioimmunotherapeutics, which involve the radiolabeling of mAbs, has emerged as a new mechanism to target cancer. A novel lutetium-177-labeled chimeric mAb developed against CD55 (177Lu-anti-CD55) was shown to reduce metastatic expansion of H460 cells in a pleural metastatic lung cancer model and improve survival in mice [174]. Interestingly, 177Lu-anti-CD55 displays synergistic anti-cancer effects when used in combination with cisplatin, significantly reducing H460 cell viability in vitro and increasing the survival of advanced pleural metastatic mice in vivo. However, on its own,177Lu-anti-CD55 appeared to be more effective than cisplatin and combinatorial treatment in early pleural metastatic models [174]. The use of bispecific antibodies in the targeting of CD55 has also recently generated interest in the treatment of cancer. With the recent successes of PD-1/PD-L1 targeting, it is unsurprising that some groups have developed bispecific antibodies targeting CD55 and PD-L1. GB262 is a novel PD-L1/CD55 bispecific antibody that has been shown to suppress PANC-1 tumor growth in mice by increasing T cell activation, as well as antibody-dependent cell-mediated cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC) [175]. Interestingly, GB262 binding and internalization also resulted in intracellular PD-L1 degradation [175]. Despite the vast evidence in the literature supporting the use of anti-CD55 antibodies as anti-cancer agents, very few clinical trials have been completed. However, several clinical trials have commenced for the human anti-idiotypic mAb (105AD7), which mimics CD55 and can be used as a vaccine to induce immune responses [176,177]. In a phase I trial in colorectal cancer patients, 105AD7 increased survival to 12 months post diagnosis of advanced disease, compared to 4 months in unimmunized patients [178]. Interestingly, despite producing anti-CD55 immune responses as an adjuvant agent, 105AD7 did not prolong survival in a double-blind colorectal cancer phase II trial [176,179]. Similar trials with 105AD7 in osteosarcoma exhibited moderate anti-cancer ability, prolonging survival in patients who mounted immune responses [177]. PAT-SC1 is an anti-CD55 IgM antibody that has undergone clinical trials. Indeed, in a gastric cancer clinical trial, a single dose of PAT-SC1 prior to resection surgery resulted in prolonged survival outcomes in a ten-year follow-up [180].

5.2. Peptides

One mechanism to investigate potential CD55-specific peptide ligands is phage display. Using this technology, Li and colleagues identified a CD55-specific ligand peptide known as CD55sp [181]. Treatment of CD55high cervical cancer cell lines, SiHa and HeLa, with CD55sp revealed inhibitory effects on cell proliferation, with IC50 values of 208.4 μg/mL and 230.3 μg/mL, respectively. Furthermore, CD55sp promoted apoptosis in SiHa and HeLa cells, evidenced by increased TUNEL staining Annexin V-PE/7-AAD flow cytometry. Recently, the targeting properties of CD55sp have been used in C-phycocyanin/carboxymethyl chitosan/CD55sp (C-PC/CMC-CD55) nanoparticles to induce apoptosis and inhibit proliferation (IC50 = 40 μg/mL) in HeLa cells [182]. In vivo, C-PC/CMC-CD55 nanoparticles effectively accumulated in HeLa tumors and significantly attenuated tumor growth compared to control. Interestingly, ELISA assays revealed that administration of C-PC/CMC-CD55 in mice increased levels of interleukin (IL)-6 and tumor necrosis factor alpha (TNFα), while reducing levels of TGFβ, suggesting at least some level of immune modulation as a possible explanation for the reduction in tumor volume observed in vivo.

5.3. Pharmacologic Inhibitors

Compared to the wealth of anti-CD55 antibodies that have been developed as anti-cancer agents, relatively few pharmacological inhibitors have been investigated. Indirect mechanisms of CD55 inhibition include the targeting of prostaglandin D2, which has been shown to induce CD55 expression in a cyclic AMP (cAMP)-dependent manner [183]. The novel indole compound AWT-489 is a prostaglandin receptor (DP) antagonist, thus, reducing the production of cAMP and CD55 [184]. The treatment of colon cancer cell line, LS174T with 10 μM of AWT-489 reduced CD55 levels [184]. However, whether AWT-489 displays significant anti-cancer effects has yet to be investigated.

5.4. Other CD55-Targeting Strategies

Additionally, inhibiting CD55 through genetic means such as CRISPR/cas9 and siRNAs has been proposed. CRISPR/cas9-knockout of CD55 in cervical cancer C33A cells suppressed their proliferative and self-renewal capabilities, sensitized cells to radiation, and decreased tumorigenicity in vivo [171]. Lipoplex-mediated delivery of siRNAs against CD55, CD46, and CD59 sensitized breast cancer BT474 and SKBR3, ovarian cancer SKOV3, and lung cancer Calu-3 cells to trastuzumab and pertuzumab [185]. Despite minimal sensitizing effects on its own in BT474, SKOV3, and Calu-3 cell lines, CD55 siRNA alone sensitized SKBR3 cells to the combined trastuzumab and pertuzumab treatment. Interestingly, siRNA-mediated inhibition of CD55 alone increased C3d deposition on all four cell lines, which is an important mediator of opsonization for macrophages. While the effect of CD55 knockdown alone on macrophage-mediated cytotoxicity was not investigated, combined CD55, CD46, and CD59 knockdown in opsonized BT474 cells significantly increased macrophage-mediated cytotoxicity [185].
In conclusion, CD55 has been shown to promote cancer stem cell characteristics in multiple forms of cancer, highlighting its significance in tumorigenesis and aggressive disease. The exploration of therapeutic strategies, particularly the development of anti-CD55 antibodies has shown promising results in preclinical studies and clinical trials. However, despite the encouraging results, challenges such as off-target effects and the pivotal role of CD55 in the complement system necessitate ongoing research to refine and optimize CD55-targeted therapies.

6. CXCR4

The chemokine C-X-C receptor 4 (CXCR4) is an essential axis in normal development and immunity. Its cognate ligand is C-X-C ligand 12 (CXCL12), also referred to as stromal cell-derived factor 1 (SDF-1). The CXCL12-CXCR4 axis is evolutionarily conserved, as knockout is embryonic lethal [186]. In the context of the immune system, CXCR4 plays an important role in the migration of lymphocytes by activating proteins involved in cell arrest and adhesion to the endothelium and migration into tissues, such as integrins [186]. In tissue injury, pro-inflammatory damage-associated molecular patterns are released, such as high mobility group box 1 (HMGB1). HMGB1 binds to CXCL12, leading to a conformational change that enhances its affinity for CXCR4, thereby enhancing the migration of immune and mesenchymal cells [187]. CXCR4 is commonly expressed on hematopoietic, endothelial, and stem cells, which will traffic to sites based on concentration gradients of CXCL12 release [188]. Importantly, the CXCR4-CXCL12 axis is responsible for the recruitment of stem cells during wound healing [189]. CXCR4 has an important role in the homing of progenitor cells to the bone marrow; thus, a blockade of CXCR4 is used when mobilization of cells to the periphery is desired (e.g., in some blood cancers like non-Hodgkin’s Lymphoma) [186].
CXCR4 tends to be aberrantly expressed in many cancer types. High CXCL12/CXCR4 may be predictive of poor prognosis in many cancer types as it is associated with higher stage and metastasis and lower survival [190,191]. Furthermore, CXCR4 stimulation by CXCL12 leads to the downstream activation of pathways like mitogen-activated protein kinases (MAPK), phosphatidylinositol-3-kinase (PI3K) pathway activation, and JAK/STAT pathways, ultimately leading to the homing of stem cells, hematopoietic development, angiogenesis, and tumor progression [186]. As CXCR4-CXCL12 is essential for cell migration during embryonic development, including organogenesis and vascularization, it could be a mechanism of inducing a less differentiated phenotype and contribute to metastasis [188]. Thus, it is unsurprising that CSCs tend to overexpress CXCR4, which permits their migration to target areas of high CXCL12 secretion. In gastric cancer, high CXCR4 expression is an indicator of poor prognosis for survival [192]. Indeed, CD44-positivity in gastric CSCs is associated with enhanced metastatic ability, as well as an ability to differentiate and recapitulate a heterogeneous tumor [193]. Similarly in prostate cancer, isolated CXCR4+ cells displayed a 3.8-fold increase in tumorigenicity, and tumor growth in vivo compared to CXCR4 cells [194]. Ovarian cancer cell lines with high CD44 expression exhibit increased resistance to cisplatin, and limiting dilution assays revealed enhanced tumorigenicity with as few as 1000 cells [195]. Similarly in breast cancer, CXCR4 was shown to maintain a tamoxifen-resistant CSC population in MCF-7 cells [196]. Thus, targeting CXCR4 is being explored as a viable option as an anti-CSC therapeutic. Drugs targeting CXCR4 are used in clinical practice for various settings, including human immunodeficiency virus and the mobilization of hematopoietic stem cells for transplantation; however, there has been a renewed focus on the targeting of CXCR4 in cancer [186,197,198]. Table 1 provides a summary of the inhibitors.

6.1. Antibodies

Several antibodies have been developed to target CXCR4 in cancer and other diseases. Ulocuplumab (BMS-936564/MDX-1338) is a human IgG4 mAb specific for CXCR4 that blocks the binding of CXCL12. In chronic lymphocytic leukemia, only CXCR4+ cell lines exhibited increased apoptosis when treated with ulocuplumab [199]. Similarly, in a preclinical study of AML, treatment with ulocuplumab resulted in an induction of apoptosis in a panel of cell lines [198]. Ulocuplumab also exhibited potent tumor growth inhibition in mouse models of lymphoma, multiple myeloma, AML and breast cancer [198,200]. Interestingly, CXCR4 overexpression in multiple myeloma cells leads to the acquisition of a mesenchymal EMT phenotype, a feature that is frequently associated with stemness [201]. Treatment with ulocuplumab inhibited the EMT phenotype in multiple myeloma tumors, decreased tumor volumes, and synergized with another standard chemotherapy, lenalidomide [201]. In a clinical setting, ulocuplumab synergizes with standard of care chemotherapies. A phase I trial of ulocuplumab combination therapy with ibrutinib in Waldenström macroglobulinemia demonstrated an overall response rate of 100%, with 33% of patients achieving a very good partial response (VGPR) [202]. Another clinical trial in multiple myeloma demonstrated that the combination of ulocuplumab with standard chemotherapies resulted in a progression-free survival rate greater than results in the standard therapy alone from previous trials, indicating that anti-CXCR4 treatment may enhance the efficacy and longevity of the standard treatment response in hematological cancers [203]. Ulocuplumab is currently being tested in clinical trials for solid and liquid cancers (NCT01120457, NCT02472977, NCT02305563, NCT03225716, NCT01359657, NCT02666209).
Other anti-CXCR4 antibodies such as PF-06747143 have demonstrated reduced cancer burden and migration and synergized with standard-of-care chemotherapies in preclinical models of hematological malignancies [204,205]. In preclinical models of chronic lymphocytic leukemia, PF-06747143 induced cell death with high specificity to cancer cells, synergized with standard of care chemotherapy drugs, and improved the survival of treated mice [206]. Additionally, PF-06747143 is significantly more effective than plerixafor, a pharmacological CXCR4 inhibitor, at reducing CXCL12-induced migration and actin polymerization [206]. PF-06747143 is in an ongoing trial (NCT02954653) but there are no results yet posted. Similarly, 12G5 is an anti-CXCR4 antibody that has demonstrated anti-tumor and anti-metastatic activity in both endometrial cancer and osteosarcoma xenografted mouse models [207,208]. Nanoantibodies such as ALX-0651 have also been investigated to target CXCR4. In a phase I study in healthy volunteers (NCT01374503), ALX-0651 demonstrated appreciable CXCR4 targeting ability; however, the study and any subsequent investigations into ALX-0651 as an anti-cancer agent were halted due to an inability to outcompete other CXCR4 antibodies [209]. Other anti-CXCR4 antibodies include LY2624587, hz515H7 (F-50067), and MEDI3185, which exhibit anti-tumor activity in hematological malignancies in mice [210,211,212]. LY2624587 (NCT01139788) and hz515H7 have both been tested in Phase I clinical trials. While results for LY2624587 have not yet been posted, hz515H7 in combination with lenalidomide and dexamethasone resulted in a 66.7% response rate in multiple myeloma patients [213].

6.2. Pharmacologic Inhibitors

Several CXCR4-antagonist small-molecule compounds have also been investigated, with Plerixafor (AMD3100, Mozobil®) being approved by the US Food and Drug Administration (FDA) in 2008 for multiple myeloma and lymphoma. Plerixafor is a first generation CXCR4 inhibitor and is the most studied and widely used CXCR4 inhibitor, with significant evidence establishing its efficacy as an anti-cancer agent in many cancer types [214,215,216]. Preclinical studies in breast, lung, colon, prostate, and pancreatic cancer showed that CXCR4high tumors have enhanced metastatic potential compared to CXCR4low, and plerixafor as a monotherapy or combined with chemotherapy successfully reduces tumor growth and metastases through inhibition of the extracellular signal-regulated kinase 1/2 (ERK1/2) and AKT pathways [214,216,217,218,219,220,221]. Zhou et al. found that CXCR4 may be a key player in tamoxifen resistance in breast cancer, as resistance is reversed when CXCR4 is knocked down or upon treatment with plerixafor due to inhibition of the AKT phosphorylation pathway [190]. In vivo, plerixafor and tamoxifen significantly reduced tumor growth compared to tamoxifen alone, with a decrease in tumor CXCR4 expression [190]. Co-culture of PC3-luc prostate cancer cells with CXCL12-producing stromal cells induces resistance to the chemotherapy drug docetaxel and protects the cells from apoptosis; however, when they are treated with docetaxel and plerixafor, they become sensitive. The combination therapy has a synergistic effect in vivo with reduced tumor burden and metastasis [221]. Treatment with plerixafor in a mouse model of metastatic breast cancer revealed that plerixafor reduced stromal cancer-associated fibroblasts—which play an important role in drug resistance and immune exclusion, reduced hypoxia, and fibrosis-related genes like Tgfβ1, while increasing the expression of activated immune-related genes like interferon gamma (IFNγ) and granzymes A and B [222]. Plerixafor synergized with anti-PD-1 and anti-CTLA-4 to improve survival and reduce metastases in mouse models of breast cancer [222]. These studies implicate CXCR4 as a key player in metastasis and drug resistance that can be overcome with CXCR4 inhibitors.
Finally, plerixafor has demonstrated specific effects on CSCs. Heckmann et al. hypothesized that the CXCR4 axis supports drug resistance by inducing dormancy associated with CSCs, therefore disrupting CXCR4 signaling with plerixafor overcomes resistance by mobilizing dormant cells from their protective niches [216]. Colon cancer cell lines with high CXCR4 are inherently more stem-like and susceptible to cytostatic drugs compared to their low expression counterparts. Addition of plerixafor increased the sensitivity of these stem-like cells to the cytostatic chemotherapy drugs tested [216]. Mobilization of quiescent stem-like cells into the periphery from their protective niches using CXCR4 antagonists should make them more susceptible to chemotherapy and aid in overcoming drug resistance.
Other CXCR4 antagonists are being generated and tested. Mavorixafor (X4P-001) is an oral allosteric CXCR4 inhibitor currently in clinical trials: NCT02823405, NCT05103917, NCT02667886, NCT02923531, and NCT04274738. In a phase I trial of clear cell renal cell carcinoma, mavorixafor treatment resulted in a significant decrease in latency-associated peptide of TGF-β1 and angiopoietin-1 from baseline [223]. Another phase I trial tested the efficacy of mavorixafor and pembrolizumab on immune cell infiltration to tumors in melanoma [224]. Combination therapy enhanced the tumor inflammatory score and supported the idea that anti-CXCR4 treatment may reduce immunosuppression in tumors. USL311 is another selective CXCR4 antagonist that is currently undergoing a clinical trial in glioblastoma (NCT02765165). A similar CXCR4 antagonist, PRX177561, in combination with anti-VEGF therapy in glioblastoma, significantly extended the time to progression and disease-free survival in several mouse models [225]. Of note, the authors found that CXCR4 is more highly expressed in glioblastoma CSCs than differentiated cancer cells, and the stem cells are more sensitive to PRX177561 compared to differentiated cells [226]. The CXCR4 antagonist MSX-122 has generated interest as the only oral non-peptide CXCR4 inhibitor. MSX-122 has displayed remarkable anti-metastatic efficacy at low doses (4 mg/kg/day) in several metastatic mouse models of breast cancer, head and neck squamous cell carcinoma, and uveal melanoma [227]. A phase I clinical trial for MSX-122 (NCT00591682) commenced in 2007; however, it was suspended for unknown reasons.

6.3. Peptides

The use of synthetic peptides has also generated interest in the targeting of CXCR4. Motixafortide (BL-8040, BKT-140, TNI4001) is a synthetic peptide CXCR4 inhibitor being tested in both solid and liquid cancers. Motixafortide binds the same pocket on CXCR4 as its ligand CXCL12/SDF-1, thereby stabilizing the inactive conformations of CXCR4, preventing its activation [228]. Since Motixafortide acts as an inverse agonist, occupying the binding pocket of CXCR4 without stimulating the receptor, it prevents the rehoming of leukemic blasts to the bone marrow. In a phase IIa trial investigating BL-8040 with high-dose cytarabine in AML, the complete response rate was 29–39% based on dosing regimen (NCT01838395) [229]. Bone marrow aspirates from patients following BL-8040 treatment demonstrated an approximately 73% decrease in CXCR4 BL-8040-occupied AML blasts from pre-treatment levels, indicating that the treatment effectively mobilized cancer cells from the bone marrow to the peripheral blood. Motixafortide induces apoptosis in AML by upregulation of miR-15a/miR-16-1, which resulted in the downregulation of antiapoptotic genes Bcl-2, Mcl-1, and cyclin D [229,230]. The mobilization of cells from the bone marrow to the periphery has important implications in the treatment of blood cancers, as the mobilization of quiescent stem-like cells into the blood stream from their protective niches using CXCR4 antagonists should make them more susceptible to chemotherapy.
Motixafortide has demonstrated success in the treatment of solid tumors. Lefort et al. used breast cancer patient-derived xenografts (PDXs) to test the efficacy of motixafortide compared to the first generation CXCR4 inhibitor, plerixafor, to decrease tumor growth and metastasis [218]. Interestingly, motixafortide demonstrated significant tumor inhibition in human-epidermal growth factor receptor 2 overexpressing (HER2+) breast PDX tumors but was not effective at inhibiting the TNBC PDXs. Furthermore, two trials have been conducted investigating motixafortide in pancreatic cancer. Due to the connection between CXCR4 and immunomodulation, the COMBAT trial (NCT02826486) found that the combination of BL-8040 and pembrolizumab therapy resulted in a rapid and significant increase in leukocytes and lymphocytes to the peripheral blood, with a notable decrease in anti-inflammatory regulatory T cells [231]. The disease control rate of combined BL-8040 with pembrolizumab was 34.5% in PDAC, with increased infiltration of CD8+ cytotoxic T cells in the tumor. The authors noted that this study did not have a chemotherapy backbone, so while the results are modest, they do suggest that BL-8040 not only enhanced pembrolizumab response in pancreatic cancer but may expand the benefits of standard chemotherapy treatment [231]. In the subsequent COMBAT trial in pancreatic cancer (NCT02826486), motixafortide and pembrolizumab were used in combination with chemotherapy. Combination with a chemotherapy regimen resulted in an overall response rate of 21.1% with a disease-control rate of 63.2% for a population of patients with poor prognosis and aggressive disease [232].
LY2510924 is a peptide antagonist of CXCR4 being tested in ongoing clinical trials: NCT02737072, NCT02652871, NCT01391130, NCT01439568. Thus far, it has demonstrated limited clinical success in tumor reduction [233,234,235]. In AML, LY2510924 caused a dose-dependent increase in mobilization of AML blasts to the peripheral blood from baseline (>20 fold-change) within one cycle of treatment [235]. A similar peptide CXCR4 antagonist, CTCE-9908, significantly reduced the tumor burden in MDA-MB-231 breast cancer tumor-bearing mice and displayed a 20-fold reduction in metastasis compared to control [236]. CTCE-9908 has been investigated in one phase I/II clinical trial for solid tumors, with the best response being stable disease [237]. More recently, the development of IS4, a novel competitive CXCR4 antagonist, reduced CXCL12-induced migration of prostate and melanoma cell lines more effectively than plerixafor [238].
These studies highlight the ongoing investigation of novel CXCR4 antagonists to treat various cancer types. In sum, CXCR4 expression is associated with worse survival and is overexpressed in many cancers. It promotes cancer by enhancing proliferation, migration, and invasion; promotes metastasis by homing mesenchymal cells to protective niches; and is immunosuppressive. There is significant clinical evidence to support the use of CXCR4 inhibitors to target these mechanisms and safely synergize with standard-of-care treatments in both hematological and solid malignancies. Next generation antagonists, such as motixafortide, are exhibiting enhanced efficacy at very low doses and are well-tolerated in clinical practice. These inhibitors also have specific anti-cancer effects on cancer stem-like cells that are key drivers of metastasis, drug resistance, and cancer recurrence.

7. CD133

CD133 (also known as prominin-1) is a pentaspan membrane glycoprotein that was first identified on murine neural stem cells and human hematopoietic stem cells but has since been found on epithelial cells in multiple tissue types [239]. CD133 is preferentially expressed on microvilli, as well as plasma membrane protrusions such as filopodia, and has also been identified as a cholesterol-interacting protein [239,240]. Interestingly, some evidence suggests that cholesterol microdomains labeled with CD133 preserve stem cell characteristics by inhibiting differentiation [241]. CD133 is also important for the proper morphogenesis of rod photoreceptor cells and has thus been suggested to play a role in proper retinal development [242]. Due to its well-appreciated role as a stem cell marker, it is unsurprising that CD133 is a well-characterized CSC marker as well [239,240]. CD133 was first used to identify CSCs in brain tumors, including glioblastoma and, later, many other cancers [240,243,244,245,246]. In prostate cancer, CD133high cells exhibit enhanced self-renewal and differentiation capabilities. Indeed, isolated CD133high cells gave rise to a phenotypically heterogeneous population in culture [247]. Similarly in liver cancer, CD133+ cells possess greater ability to form tumor spheroids in vitro and exhibited enhanced tumorigenicity in vivo [248]. Only CD133+ cells sorted from liver tumor xenografts implanted into secondary mouse recipients generated tumors, and tumor growth was enhanced in the secondary tumors compared to primary tumors [248]. In colorectal cancer, CD133+ cells isolated from surgically resected tumor specimens generated long-term spheroid cultures that displayed self-renewal and differentiation capabilities, eventually recapitulating the original tumor phenotype [249]. Furthermore, the CD133+ spheroid cells were resistant to irinotecan, a further indication of chemotherapy resistance associated with stemness [249]. CD133 has also been identified as a CSC marker in lung cancer. Patient-derived CD133+ lung cancer spheroids displayed enhanced tumorigenicity in vivo, and immunohistochemical analysis revealed that the subsequent CD133-derived tumors reproduced a heterogeneous phenotype resembling that of the original patient tumor [250]. High CD133 expression is associated with enhanced clonogenicity, invasiveness, and resistance to chemotherapy in oral squamous cell carcinoma [251]. Clinically, CD133-positivity is associated with aggressive disease and poor prognosis in colorectal and invasive breast cancer [252,253]. However, the role of CD133 as a CSC marker is not without controversy. In renal cell carcinoma, high CD133 expression is associated with prolonged survival and is a marker for good prognosis [254]. Conversely in pancreatic adenocarcinomas, CD133-positivity does not confer significantly enhanced tumorigenicity compared to other CSC markers [255]. Thus, the role of CD133 as a CSC marker is context- and tumor tissue-type dependent. Regardless, given the role of CD133 as a robust CSC marker across multiple different malignancies, targeting CD133 presents an interesting avenue to disrupt stemness and improve patient outcomes (summarized in Table 1).

7.1. Antibodies

Several anti-CD133 mAbs have been developed, with many serving as conjugates to nanoparticles. Indeed, conjugation of an anti-CD133 mAb to PEGylated gold nanoparticles was used to target 5-FU delivery in colorectal cancer HCT116 cells [256]. Similarly, targeted paclitaxel delivery to breast cancer MCF-7 cells with anti-CD133 mAb targeting nanoparticles significantly reduced colony-forming potential and mammosphere formation compared to paclitaxel alone [257]. Two injections of the CD133-targeting paclitaxel nanoparticles resulted in a 70% decrease in MDA-MB-231 tumor volume [257]. Another study shows that improving the targeted delivery of the topoisomerase inhibitor SN-38 by incorporating it into PEG–PCL-based nanoparticles with anti-CD133 mAb increases the cytotoxic effects of SN-38 toward colorectal cancer cells and tumor xenografts [258]. Other groups have combined antibody and cellular immunotherapies, generating bispecific anti-CD133/CD3 antibodies bound to cytokine-induced killer cells (BsAb-CIK) [259]. Intraperitoneal injections of BsAb-CIK in pancreatic SW1990-tumor-bearing mice resulted in a significant decrease in tumor growth. This finding suggests that antibody-based targeting of CD133 may hold promise in developing targeted immunotherapies. Indeed, an Fc-optimized CD133 mAb, known as 293C3-SDIE, induces NK cell activation, degranulation, secretion of IFN-γ, and antibody-dependent cellular cytotoxicity, resulting in potent lysis of colorectal cancer cells Caco-2 and HCT-116, as well as B cell acute lymphoblastic leukemia cells, highlighting its potential in solid and hematologic malignancies [260,261].

7.2. CAR-T and NK Cells

Other mechanisms of targeting CD133 include modulation of immune cells such as T and NK cells. Indeed, CAR-based therapies targeting CD133 have been developed in recent years. Anti-CD133-CAR expressing NK92 cells displayed specificity against patient-derived CD133+ ovarian cancer cells, resulting in reduced cell viability [262]. Furthermore, the co-incubation of cisplatin and anti-CD133-CAR NK92 cells on ovarian cancer cells resulted in increased cell death compared to cisplatin alone, implicating CD133-directed NK cell killing as a possible adjuvant agent for cisplatin-based regimens. More recently, anti-CD133 CAR-T cells have been tested in vivo in small cell lung cancer models (SCLC) [263]. While adoptive transfer of the anti-CD133 CAR-T cells alone reduced tumor burden and improved survival in SCLC mouse models, combined anti-PD-1 therapy, CD73 inhibition, and CD133 CAR-T cells resulted in complete remission for 25% of the mice [263]. These findings add further evidence, suggesting that combination therapy is most effective at targeting heterogenous tumor populations. At least two clinical trials for anti-CD133 CAR-T cells have been completed. Indeed, the findings of a phase I clinical trial (NCT02541370) indicated that infusion of autologous CAR-T-cell-directed CD133 (CART-133) inhibits disease progression in patients with advanced metastatic lung, liver, and colorectal carcinoma [264]. Another phase II clinical trial (NCT02541370) using anti-CD133 CAR-T cells in hepatocellular carcinoma resulted in a median progression-free survival (PFS) of 6.8 months and overall survival of 12 months; however, only one patient achieved partial response [265].

7.3. BiKEs and Vaccines

Bispecific killer engagers (BiKEs) have also been used as anti-CD133 agents in cancer. One such BiKE combines CD16 and CD133 single chain variable fragments (scFv) to engage NK cells (16 × 133 BiKE) [266]. Indeed, 16 × 133 BiKE was shown to activate resting NK cells to induce IFN-γ production and degranulation against CD133+ colorectal cancer Caco-2 cells, which are known for being resistant to NK-mediated killing. Trispecific (CD16, IL-15, CD133) and tetraspecific (CD16, IL-15, EpCAM, CD133) NK cell engagers have also demonstrated appreciable anti-cancer efficacy against colorectal Caco-2 cells, as well as a panel of breast, prostate, HNSCC, CRC, and AML cell lines, respectively [267,268]. Interestingly, a dendritic cell vaccine against CD133 was tested in a phase I clinical trial (NCT02049489) for recurrent glioblastoma [269]. In this trial, autologous dendritic cells were loaded with CD133 antigen, and while the results indicated that ICT-121 is safe and induces immune responses, it is unclear whether this treatment provided any anti-tumor benefit.

7.4. Immunotoxins

Like many other anti-CSC targeted therapeutics, immunotoxins have also played a role in CD133-targeted therapy. GMI, which is a fungal immunomodulatory protein derived from Ganoderma microsporum, reduces CD133 protein expression via autophagic-lysosomal protein degradation [270]. Indeed, oral gavage of GMI demonstrated remarkable inhibition of tumor growth in lung pemetrexed-resistant CD133+ A549/A400 tumor-bearing mice and significantly decreased CD133 expression in the tumor, indicating good tumor-penetrating capabilities. Other groups have conjugated anti-CD133 scFv to a truncated form of Pseudomonas exotoxin A (PE38), generating the novel immunotoxin dCD133KDEL [271]. dCD133KDEL has been tested against several forms of cancer, including HNSCC, breast, and ovarian, demonstrating appreciable anti-tumor effects [271,272,273]. Pre-treatment of HNSCC CD133+ UMSCC-11B cells with dCD133KDEL significantly reduced tumorigenicity in xenotransplantation mouse models, suggesting eradication of the aggressive CD133+ CSC population [271]. Bostad et al. reported success using an anti-CD133 mAb conjugated to saporin, a plant-derived toxin in the ribosome inactivating protein family [274]. Interestingly, this group used photochemical internalization to deliver AC133-saporin to WiDr colon cancer tumor-bearing mice, resulting in a significant decrease in tumor growth compared to control [274].

7.5. Pharmacologic Inhibitors

Several pharmacologic inhibitors, including small molecule inhibitors and repurposed drugs, have generated interest as CD133 inhibitors. Celecoxib, which is a non-steroidal anti-inflammatory drug, inhibits the expression of CD133 in HT29 and DLD1 colorectal cancer cells via suppressing the Wnt signaling pathway [275]. Conversely, other studies have suggested that celecoxib-induced downregulation of CD133 is mediated by AKT inhibition in HT29 cells [276]. Trifluridine, a drug that is currently in clinics for the management of refractory colorectal cancer inhibits spheroid formation in CD133+ DLD1 colorectal cancer cells, suggesting a potential mechanism for its anti-cancer activity [277]. ACT001, a guaianolide sesquiterpene lactone, inhibits stemness and CD133 expression by inducing Olig2 ubiquitination degradation in A549 and NCI-H820 lung cancer cells [278]. However, the exact anti-cancer mechanism of ACT001 remains controversial, as it was identified as a direct inhibitor of plasminogen activator inhibitor-1 (PAI-1) and exerted anti-cancer effects in glioma cells by inhibiting the PAI-1/PI3K/AKT pathway [279]. Regardless of the exact mechanism, ACT001 was approved as an orphan drug by the FDA in 2017, and phase I and II clinical trials have commenced in glioblastoma, with at least one patient achieving complete remission (ACTRN12616000228482, NCT05053880) [280].

7.6. Aptamers and Peptides

Aptamers and peptides have also been studied to target CD133. Several CD133-specific aptamers have been used as agents to deliver chemotherapeutics such as doxorubicin. Indeed, CD133 aptamer-guided delivery of doxorubicin results in a higher doxorubicin concentration in Huh7 hepatocellular carcinoma stem cells, resulting in a significant decrease in tumorsphere formation [281]. Other doxorubicin-loaded CD133 aptamers have been tested in vivo, resulting in a significant reduction in diethylnitrosamine-induced liver cancer tumors in mice [282]. Other groups have used CD133 targeted RNA aptamers to deliver an anti-microRNA (miRNA) against miR-21, a well-known miRNA involved in TNBC progression [283]. CD133 aptamer-mediated delivery of anti-miRNA21 into MDA-MB-231 breast cancer tumors in mice resulted in accumulation in the tumor, as well as a significant reduction in tumor growth [284]. A CD133-specific peptide, LS-7, was identified in 2012, which reduced migration and invasion of murine colon and breast cancer cell lines CT-26 and MA-782, respectively [285]. While LS-7 was able to localize to CD133+ CT-26 tumors in vivo, its ability to reduce tumor growth has not yet been investigated.

7.7. Other CD133-Targeting Strategies

CRISPR/cas9 and siRNAs have also been investigated as a tool to inhibit CD133 expression. Li et al. demonstrated that CRISPR-Cas9 mediated CD133 knockout in colorectal cancer LoVo cells reduced proliferation, colony formation, and EMT, as well as migration [286]. siRNA-mediated suppression of CD133 not only sensitized HT-29 cells to oxaliplatin but also decreased colony formation, migration, and expression of EMT-related genes [287]. In 2016, a study evaluated the effect of CD133 (AC133) mAb-based NIR-PIT on CD133+ U251 glioma tumors [288]. In established subcutaneous human xenograft tumors, AC133 NIR-PIT demonstrated significant tumor growth inhibition in CD133-expressing U251 glioma models, confirmed by reduced signal intensity post-NIR light exposure and diminished tumor volumes. Western blot analyses of tumor cell lysates revealed a substantial reduction in CSC markers, indicating specific killing of undifferentiated CSCs and subsequent tumor growth inhibition, as evidenced by various imaging modalities and tumor weight measurements [288].
CD133 serves as a pivotal marker for CSCs across various malignancies, correlating with aggressive disease and poor prognosis in several cancers such as glioblastoma, colorectal, breast, lung, and liver cancers. Various strategies have been explored to target CD133, including antibody-based therapies, CAR-based immunotherapies, pharmacologic inhibitors, immunotoxins, and RNA-based approaches, each showing promising results in preclinical and clinical studies. Further research and clinical trials are warranted to fully explore the therapeutic potential of CD133-targeted interventions and their impact on improving patient outcomes across diverse malignancies.

8. Nanog

Nanog is a DNA binding Homeobox transcription factor essential for maintaining pluripotency and self-renewal. Nanog is mainly expressed in pluripotent and developing germ cells and is essential in maintaining stemness (undifferentiated state of the cells), as well as determining embryonic cell fate [289]. Nanog is epigenetically switched off during development. However, in cancers, the Nanog gene is broadly expressed in many cancer types, especially of the epithelial phenotype [290,291,292,293]. Nanog-mediated oncogenic reprograming is observed in malignant cancers and is associated with clinical manifestations of metastasis and secondary tumors [290,292,293,294]. It is well-appreciated that Nanog expression confers a CSC phenotype across multiple cancer types, including liver, colorectal, prostate, breast, ovarian, lung, and brain [293]. Indeed, a meta-analysis of the prognostic significance of elevated Nanog expression in solid tumors revealed that it is associated with more aggressive disease and worse prognosis [295]. Nanog expression is associated with tumorsphere and colony formation, as well as chemoresistance and tumorigenicity in breast cancer [296]. In colorectal cancer, enforced Nanog expression was shown to promote tumorigenicity and generate tumors in mice [297]. Furthermore, Nanog has been identified as a CSC marker in both prostate and HCC cancers [290,298]. Nanog can also modulate expression of other CSC markers, including ALDH, CD44, and CD133, further cementing its role as an important mediator of stemness [290,299,300,301]. Collectively, this makes Nanog a lucrative target to treat advanced cancers.

8.1. PMOs and RNA Interference

To date, no drugs that directly target Nanog have been identified, with most chemical inhibitors having a secondary effect of Nanog reduction. Some functional drugs using synthetic DNA and RNAi machinery that directly target NANOG have been identified. Kundu et al., 2020, used synthetic DNA antisense phosphorodiamidate morpholino oligonucleotides (PMOs) targeting NANOG in MCF-7 cells [302]. PMOs are complimentary to the gene start site and block transcription through steric blocking [303]. However, PMOs do not enter intact cells, making their delivery challenging, needing carrier molecules like cell penetrating peptides, nanoparticles, or dendrimers [304]. In this study, the anti-Nanog PMO was conjugated to a nonpeptidic internal guanidinium transporter (IGT) to facilitate intracellular delivery [302]. The IGT-PMO decreased Nanog protein expression in breast cancer MCF-7 cells. Further analysis revealed that Nanog inhibition also reduced clonogenicity, as well as migration and invasion potential, and sensitized MCF-7 cells to Taxol chemotherapy. However, it is important to note that CXCR4 and MMP9 were also downregulated as the result of Nanog inhibition and, thus, may be responsible for the reduction observed in migration and invasion [302]. Nanog knockdown with siRNA has also been shown to increase the sensitivity to 5-FU and decrease self-renewal as well as migration in of CRC SW-480 cells [305].

8.2. Pharmacologic Inhibition

Indirect Nanog inhibition is achieved by drugs targeting upstream regulators or those that mediate proteasomal degradation of Nanog. Histone deacetylase (HDAC) has previously been shown to regulate Nanog expression in embryonic stem cells [306]. Suberoylanilide hydroxamic acid (SAHA), which is an FDA-approved drug for T-cell lymphoma as a pan-HDAC inhibitor, markedly reduced Nanog expression in cisplatin-resistant HNSCC cells, sensitizing them to cisplatin [307,308]. In vitro, SAHA reduced CAL27-CisR and UD-SCC-2-CisR tumorsphere formation, and in vivo, significantly reduced tumor growth and metastasis [308]. In both cases, Nanog expression was significantly downregulated. PiB (diethyl-1,3,6,8-tetrahydro-1,3,6,8-tetraoxobenzo [lmn]3, 8 phenanthroline-2,7-diacetate) has also been investigated as an indirect Nanog inhibitor in cancer [309]. PiB is a well-characterized peptidyl-prolyl-cis-trans isomerase NIMA-interacting 1 (Pin1) inhibitor [310,311,312]. Pin1 has been identified as an upstream Nanog regulator by inhibiting speckle-type POZ protein (SPOP)-mediated Nanog polyubiquitination and degradation [309,310]. Indeed, Pin1 inhibition by PiB degrades Nanog though ubiquitination in prostate cancer, resulting in decreased tumorsphere formation and colony growth [309]. Similarly, resveratrol, a phytochemical that has been investigated as an anti-cancer agent in multiple cancer types, reduced Nanog protein levels in glioblastoma by activating proteasomal degradation [313,314]. Treating glioblastoma CSCs with resveratrol followed by intracranial injection into nude mice significantly reduced tumorigenicity and prolonged survival compared to control mice [313]. Interestingly, Nanog has also been identified as a downstream target of aspirin, a well-known non-steroidal anti-inflammatory drug [315]. Indeed, aspirin decreased Nanog protein levels in colorectal cancer HCT116 and LoVo cells by decreasing Nanog stability in a proteasome-independent manner. Consequently, aspirin-treated HCT116 and LoVo cells displayed attenuated stemness features, including tumorsphere formation [315]. Metformin is an FDA-approved first-line drug for diabetes mellitus that has been shown to downregulate Nanog through upstream reduction of kruppel-like factor (KLF5) levels in TNBC [316]. Indeed, metformin suppressed TNBC CSCs at least partially by inhibiting KLF5, evidenced by reduced tumorigenesis in HCC1806 cells implanted into nude mice. In esophageal cancer, iron chelators, DFX and SP10, appeared to suppress the expression of stemness markers, including Nanog [317]. In vivo, SP10 significantly reduced Nanog expression in TE8 tumors and decreased tumor growth.
Thus, Nanog dysregulation in cancer contributes to oncogenic reprogramming, maintenance of pluripotency and self-renewal, and aggressive tumor behaviors. Despite the lack of direct Nanog-targeting drugs, promising avenues for therapeutic intervention have been explored. From synthetic DNA and RNAi approaches to indirect inhibition through targeting upstream regulators or mediators of proteasomal degradation, diverse strategies have shown efficacy in attenuating Nanog expression and mitigating CSC characteristics across various cancer types.

9. Notch

Notch1, 2, 3, and 4 are evolutionarily conserved genes involved in various biological processes, including organ development and repair and hematopoietic stem cell differentiation and development [318]. The Notch genes encode four receptors that bind to notch ligands (delta-like 1,4 and jagged 1,2) present in adjacent cells [319]. Upon activation, the notch receptors are cleaved by ADAM family metalloproteases or gamma-secretase, then the notch intracellular domain (NICD) is internalized to the nucleus, resulting in transcriptional regulation [318]. Canonical Notch signaling is different from other receptor mediated signaling as it does not have signaling intermediates [320,321]. While Notch has been identified as a tumor suppressor in some cancers, it has also been implicated as an important contributor to maintaining stemness in CSCs [318]. Indeed, Notch signaling maintains the CSC population in patient-derived glioma cells, evidenced by increased tumorsphere formation and an ability to self-renew and differentiate, effects which were abrogated upon Notch inhibition [322]. In renal cell carcinoma, Notch signaling was upregulated in CD24+/CD133+ CSCs, and their stemness was at least in part due to Notch activation and signaling, highlighting the role of Notch in promoting expression of other CSC markers [323]. Similarly, Notch expression was upregulated in CD44+/CD24+/ESA+ pancreatic CSCs [324]. Notch pathway inhibition in this CSC population significantly attenuated tumorsphere formation and tumorigenicity in mice [324]. Notch reporter systems have also identified Notch as a major contributor to CSC populations in breast cancer. Indeed, Notch+ cells were able to generate heterogeneous Notch+ and Notch SUM159 cells, an effect that was not observed in Notch cells [325]. Moreover, Notch+ SUM159 and MCF-7 cells exhibited increased tumorigenicity in limiting dilution xenograft assays. Thus, targeting Notch signaling presents a promising therapeutic strategy (Figure 2, Table 1).

9.1. Pharmacologic Inhibitors

One of the major mechanisms of chemical Notch inhibition is gamma-secretase inhibitors (GSI). Indeed, GSIs are well-studied for Notch intracellular domain (NICD) internalization resulting in notch inhibition. However, one of the pitfalls of Notch inhibition with GSIs is that this class of drugs exhibit significant gastrointestinal toxicity, which limits their clinical translatability. Regardless, several GSIs have been developed, including crenigacestat (LY3039478), LY900009, osugacestat (AL101, BMS-906024), RO4929097 (RG473), and nirogacestat (PF-03084014) [326]. Crenigacestat is a small molecule first generation GSI exhibiting potent NICD inhibition, with a reported IC50 of approximately 1 nM in vitro [327]. Several preclinical studies have demonstrated that crenigacestat is a robust anti-cancer agent with oral bioavailability against breast, colon, lung, ovarian, glioblastoma, gastric, and intrahepatic cholangiocarcinoma PDX models in mice [327,328]. Due to its preclinical success, several clinical trials have been completed investigating crenigacestat in humans. A phase I trial of crenigacestat monotherapy (NCT01695005) in patients with advanced metastatic cancer resulted in modest anti-cancer effects, with one breast cancer patient achieving partial response and ~31–35% of patients achieving stable disease [329]. Other clinical trials have shown that crenigacestat exhibits limited clinical activity in adenoid cystic adenocarcinoma (NCT01695005) and advanced solid tumors (NCT02836600) [330,331]. Interestingly, B cell maturation antigen (BCMA) directed CAR-T cells in combination with crenigacestat appears to demonstrate appreciable anti-cancer efficacy in patients with multiple myeloma (NCT03502577) [332]. LY900009 is a similar GSI that inhibits NICD cleavage and displays an IC50 of 0.005–20 nM [333]. However, in a phase I clinical (NCT01158404) trial for advanced cancer, no patients experienced appreciable response to LY900009 beyond stable disease [333]. Osugacestat (BMS-906024, AL101) is a benzodiazepine succinimide GSI with pan-Notch inhibition activity [334]. Potent anti-tumor activity in preclinical testing with osugacestat against breast MDA-MB-468, T-acute lymphoblastic leukemia (T-ALL) and adenoid cystic carcinoma mouse models led to the design of several phase I and II clinical trials [334,335]. In a phase I trial in patients with relapsed T-ALL, 32% of patients displayed a 50% reduction in bone marrow blasts, with two patients achieving CR and PR, respectively [336]. More recently, the results of the ACCURACY trial (NCT03691207) for osugacestat in recurrent/metastatic adenoid cystic carcinoma revealed that 12% of patients experienced partial response, while 57% achieved stable disease [337]. Osugacestat also showed synergistic activity with paclitaxel in NSCLC xenografts, and a phase I trial (NCT01653470) for osugacestat in combination with paclitaxel, 5FU+ irinotecan, or carboplatin + paclitaxel has been completed for patients with metastatic solid tumors; however, results have yet to be released [338]. R04929097 is another notch GSI with an IC50 of 4 nM [339]. RO4929097 reduced Notch signaling in melanoma and significantly reduced tumor volume as well as tumorigenicity in serial xenotransplantation models [340]. Phase II clinical trials in metastatic colon cancer (NCT01116687), melanoma (NCT01120275), sarcoma (NCT01154452), and pancreatic adenocarcinoma (NCT01232829) showed limited clinical efficacy [341,342,343,344]. Nirogacestat (PF-03084014) is a GSI with an IC50 of 6.2 nM. Preclinical studies revealed that nirogacestat has anti-CSC properties, reducing self-renewal and proliferation of HCC CSCs and exhibiting potent anti-tumor activity [345]. Interestingly, nirogacestat appears to synergize with docetaxel in prostate cancer, significantly reducing tumor growth in mice, as well as the frequency of CSCs in the tumors [346]. Numerous phase I and II clinical trials have been completed for nirogacestat across multiple cancer types. In a phase II trial (NCT01981551), nirogacestat monotherapy for pre-treated patients with dermoid tumors resulted in a 29% confirmed partial response rate [347]. More recently, phase II (NCT04195399) and phase III (NCT03785964) trials for nirogacestat in desmoid tumors in pediatric and adult populations, respectively, have commenced. Preliminary results from the phase III trial indicate an objective response rate of 41% compared to the placebo [348]. Clinical trials for nirogacestat in combination with docetaxel in patients with advanced TNBC have also been completed, with 16% of patients achieving confirmed partial response and 36% achieving stable disease (NCT01876251) [349].
Other chemical GSIs include DAPT, MRK-0752, and MRK-560. DAPT has been investigated in preclinical studies. DAPT increased the efficacy of cisplatin in osteosarcoma by directly targeting CSCs, resulting in significant tumor control in vivo [350]. DAPT also reduced CD44+ cells in gastric cancer and subsequent Wnt/β-catenin signaling, effectively inhibiting tumorsphere formation [351]. DAPT was effective in reducing Notch levels and affecting growth hormone production and tumor growth in growth-hormone-producing adenomas [352]. MK-0752 was originally developed as a treatment for Alzheimer’s disease, targeting the amyloid precursor protein, and has an IC50 of 50 nM [326]. In breast cancer MC1 xenograft models, MK-0752 reduced the CD44+/CD24 and ALDH+ CSC populations, and serial transplantation confirmed a reduction in tumorigenicity [353]. In a phase I clinical trial of MK-0752 in combination with docetaxel in breast cancer patients, 45% of the evaluable patients achieved a partial response; however, as there was only one arm in this study, it is unclear whether the efficacy was due to docetaxel or MK-0752 [353]. MK-0752 has also been investigated in preclinical models of ovarian cancer, significantly inhibiting ovarian xenograft tumor growth following cisplatin treatment [354]. Phase I and II clinical trials with MK-0752 in advanced solid tumors, PDAC (NCT01098344), and CNS malignancies have been completed with varying degrees of efficacy [355,356,357]. While no measurable responses were observed in pediatric patients with CNS malignancies, MK-0752, in combination with gemcitabine in PDAC, did result in tumor control activity, with 13 patients achieving stable disease and one achieving a confirmed partial response [356,357]. MRK-560 is a second generation GSI that reportedly displays reduced GI toxicity [358]. Indeed, treatment of T-ALL-patient-derived xenografts with MRK-560 significantly attenuated the leukemia burden and improved survival outcomes in mice without any associated GI toxicity [358].
Several other small molecule Notch inhibitors have also been developed. Limantrafin (CB103) is a recently developed small molecule inhibitor that targets the Notch transcription activation complex [359]. In TNBC and T-ALL xenograft models, limantrafin markedly reduced tumor burden and prolonged survival [359]. Limantrafin in combination with paclitaxel or fulvestrant also potently inhibited mammosphere formation in breast MCF-7 and estrogen receptor positive (ER+) cells [360]. Moreover, limantrafin synergized with paclitaxel in TNBC HCC1187 xenografts, resulting in reduced tumor growth and delayed tumor recurrence after discontinuation of treatment compared to paclitaxel alone [360]. ZLDI-8 is a novel a disintegrin and metalloproteinase 17 (ADAM-17) inhibitor, which is an enzyme involved in Notch activation and cleavage [361]. Interestingly, ZLDI-8 inhibited the EMT phenotype of NSCLC, CRC, and HCC cells, sensitizing them to chemotherapeutic agents, as well as suppressing metastasis and tumor growth in vivo [361,362,363].

9.2. Antibodies

Antibody-based notch inhibition either targets notch ligands or receptors. The mAb 602.101 is specific to Notch1 and inhibited Notch signaling in breast cancer cell line MDA-MB-231 [364]. Furthermore, treating MDA-MB-231 cells with 602.101 decreased the CD24/CD44+ CSC population and significantly attenuated primary, secondary, and tertiary mammosphere formation. Preclinical models using an antibody that targets the negative regulatory region of Notch 1 resulted in the effective control of tumor growth in Calu-6 and HM7 xenograft models [365]. Tarextumab (OMP-59R5) targets Notch receptors 2/3 and displays anti-cancer activity against several epithelial cancers, including breast, lung, ovarian, and pancreatic [366]. Limiting dilution assays derived from tarextumab-treated pancreatic PN8 xenograft tumors revealed that tarextumab reduces CSC frequency and tumorigenicity. Furthermore, tarextumab synergized with paclitaxel and gemcitabine to markedly inhibit tumor growth in PN8 xenograft models [366]. Phase I clinical trials with tarextumab (NCT01859741) showed an 84% overall response rate in small cell lung cancers in combination with etoposide and platinum; however, this treatment did not improve PFS, and the trial was thus terminated [367]. While tarextumab monotherapy in patients with solid tumors (NCT01277146) resulted in reduced Notch signaling, no evaluable responses beyond stable disease were observed [368]. Similarly, a randomized phase II trial (NCT01647828) of tarextumab in combination with nab-paclitaxel or gemcitabine did not improve PFS or overall survival in patients with metastatic pancreatic cancer [369]. Brontictuzumab (OMP-52M51) is a Notch1 antibody that has been investigated in several clinical trials. In a phase I trial for solid tumors (NCT01778439), clinical benefit was observed in 17% of patients [370]. In hematologic malignancies, brontictuzumab displayed moderate anti-tumor activity (NCT01778439) [371]. Brontictuzumab has also been investigated in phase I trials for metastatic CRC (NCT03031691), adenoid cystic carcinoma (ACC) (NCT02662608), and lymphoid malignancies (NCT01703572). In 2019, a phase I clinical trial (NCT02129205) with a novel anti-Notch3 antibody-drug conjugate with auristatin, PF-06650808, 50% of breast cancer patients achieved stable disease [372]. Interestingly, among ER+ patients, 21% achieved a partial response.
Several anti-DLL4 antibodies have also been developed to target Notch. Enoticumab (REGN421) is an anti-DLL4 mAb that displayed modest anti-tumor activity in phase I clinical trials (NCT00871559) for patients with solid tumors, with 50% of patients having SD, and 2 out of 32 achieving PR [373]. Demcizumab (OMP-21M18) is another anti-DLL4 antibody, which had an overall response rate of 21% in a phase Ib trial (NCT01952249) for patients with ovarian, peritoneal, and fallopian tube cancer when combined with paclitaxel [374]. In a similar trial for NSCLC (NCT01189968), 50% of patients had objective tumor responses when treated with demcizumab in combination with pemetrexed and carboplatin [375]. Several patients experienced significant cardiac toxicity; however, this effect could be attenuated with a truncated dosing regimen. Other clinical trials for demcizumab in combination with various anti-cancer drugs have been completed, including folinic acid, 5-FU, and irinotecan (FOLFIRI) in advanced pancreatic cancer (NCT01189942), pembrolizumab in advanced solid tumors (NCT02722954), and paclitaxel in platinum-resistant ovarian cancer (NCT01952249). MEDI0639 is a novel DLL4 antibody that inhibits angiogenesis in preclinical models [376]. However, a phase I clinical trial (NCT01952249) with MEDI0639 in patients with advanced solid tumors revealed very modest preliminary clinical results, with only 1 out of 20 patients having PR [377]. To our knowledge, no further clinical trials have been completed. In 2016, Xu et al. described a novel DLL4 antibody, MGZ01, which reduced the CD24/CD44+ breast cancer CSC population and significantly attenuated tumor growth in MDA-MB-231 xenograft models [378]. Other groups have investigated dual targeting of DLL4 and VEGF with bispecific antibodies such as navicixizumab (OMP-305B83) and ABT-165. Indeed, navicixizumab has been evaluated in phase I clinical trials for patients with solid tumors (NCT02298387) [379]. However, the anti-tumor efficacy of navicixizumab was modest, with 6.1% achieving partial response, 25.8% achieving stable disease, and 57.6% experiencing progressive disease [379]. In a separate phase I trial (NCT03030287), navicixizumab in combination with paclitaxel in patients with ovarian, peritoneal, or fallopian tube cancers resulted in an overall response rate of 43.2% [380]. In preclinical models, ABT-165 induced significant tumor growth inhibition in glioblastoma and colon cancer xenograft models [381]. A phase I trial (NCT03368859) of ABT-165 combined with FOLFIRI for patients with second-line CRC showed modest anti-tumor activity, with 3 of 16 patients displaying a partial response [382].

9.3. T-Cell Engagers

Other groups have focused on targeting notch ligands such as the Delta-like ligands (DLL). Tarlatamab (AMG 757) is a DLL3 and CD3 BiTE facilitating T-cell-mediated lysis that displayed excellent anti-tumor activity in preclinical PDX SCLC models [383]. In phase I and II clinical trials for SCLC, tarlatamab displayed promising preliminary results [384,385]. In the phase I trial (NCT03319940), 23 out of 107 patients achieved partial response, while 2 achieved complete response [384]. Similarly, in the phase II trial (NCT05060016), objective response was observed in 40% of SCLC patients [385]. A similar anti-DLL3, anti-CD3, anti-albumin T-cell engager, HPN328, is currently in a phase I/II clinical trial for patients with SCLC and other neuroendocrine cancers (NCT04471727) [386]. The preliminary results indicate moderate tumor control, with 40% of patients experiencing > 30% tumor shrinkage and at least one confirmed partial response.

9.4. Antibody Drug Conjugates

Rovalpituzumab terisine (Rova-T) is an antibody–drug conjugate that targets DLL3 [387]. While preclinical models in SCLC were promising, phase II (NCT02674568) and III (NCT03033511) clinical trials failed to reach their efficacy end points [388,389,390].
While targeting Notch signaling pathways, particularly through pan-Notch inhibitors like GSIs, presents a promising therapeutic strategy in cancer treatment, significant challenges remain. The dose-limiting gastrointestinal toxicities associated with GSIs have hindered their clinical translatability despite their demonstrated efficacy in preclinical studies. Furthermore, the limited clinical success observed across various trials highlights the complexity of Notch signaling in cancer and the need for further research to optimize therapeutic approaches. Nevertheless, the diverse array of small molecule inhibitors and antibody-based therapies targeting Notch receptors and ligands offer continued avenues for exploration in the treatment of various malignancies.

10. Wnt/β-Catenin

The Wingless-type (Wnt)/β-catenin signaling is a highly conserved pathway governing cell proliferation, survival, migration, differentiation, tissue homeostasis, and self-renewal [391,392]. The Wnt/β-catenin pathway is complex and has been extensively reviewed elsewhere [391,392,393]. There are four major segments in the Wnt/β-catenin pathway, including extracellular signals derived from Wnt proteins, cell membrane receptors such as Frizzled (FZD) and low density lipoprotein receptor-related proteins 5 and 6 (LRP5/6), cytoplasmic β-catenin and disheveled (DVL), and nuclear translocation of β-catenin to modulate expression of target genes through T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) transcription factors [391]. Posttranslational modification of Wnts such as lipid modification through porcupine O-acyltransferase (PORCN) are required for Wnt activation and ligand activity [394]. Active Wnt ligands bind to the FZD-LRP5/6 receptor complex, which, in turn, recruits and activates Dvl to disrupt the β-catenin destruction complex and consists of Axin, adenomatous polyposis coli (APC), glycogen synthase kinase 3β (GSK-3β), and casein kinase 1 (CK1). Cytoplasmic β-catenin is thus stabilized and translocates to the nucleus to interact with TCF/LEF transcription factors to induce the expression of target genes [391,393]. The Wnt/β-catenin pathway is often aberrantly expressed in both cancer and diseases such as atherosclerosis, idiopathic pulmonary fibrosis (IPF), and amyotrophic lateral sclerosis (ALS) [395,396,397]. In cancer, dysregulated Wnt/β-catenin signaling is often due to mutations in one or more components of the Wnt signaling pathway [391]. It is well-appreciated that Wnt/β-catenin signaling is important for maintaining the CSC population in cancer. For example, colon CSCs are characterized by high Wnt expression, with significantly enhanced clonogenicity, as well as tumorigenicity, in limiting dilution assays in mice [398]. In T-ALL Lin−CD3+c-KitMid cells, β-catenin overactivation is associated with expansion of the CSC population [399]. Similarly, ALDH1+ and CD24/CD44+ breast CSCs exhibited increased Wnt/β-catenin signaling activity, and Wnt knockdown or inhibition significantly reduced the tumorsphere formation associated with CSCs [400]. Moreover, Wnt/β-catenin signaling was shown to maintain self-renewal and tumorigenicity in HNSCC CSCs by promoting Oct4 expression [401]. Targeting the Wnt/β-catenin pathway is, thus, an attractive avenue for anti-cancer agents, and numerous agents have been developed to target this pathway from multiple angles (Figure 3, Table 1).

10.1. Antibodies

Antibodies targeting various components of the Wnt/β-catenin have been developed and tested against multiple forms of cancer, with many entering clinical trials. Vantictumab (OMP-18R5) is an mAb that binds to FZD1, FZD2, FZD5, FZD7, and FZD8 receptors to inhibit Wnt3a signaling [402]. In colon C28, breast UM-T3, lung Lu24, and pancreatic PN8 xenograft models, vantictumab significantly reduced tumor growth and markedly reduced tumorigenicity in subsequent limiting dilution assays in pancreatic and breast cancer models [402]. Due to its success in preclinical models, vantictumab has progressed to phase I clinical trials; however, its clinical translatability has been limited due to significant bone toxicities associated with Wnt inhibition [403,404,405]. Vantictumab monotherapy in a phase I trial for patients with advanced solid tumors resulted in prolonged SD in 3 out of 18 patients (NCT01345201) [403]. In a 2019 phase I clinical trial for metastatic pancreatic cancer (NCT02005315), partial response was documented in 42% of patients, with an additional 11% achieving stable disease when treated with vantictumab in combination with nab-paclitaxel and gemcitabine [404]. However, the study was terminated due to a failure to reach satisfactory therapeutic index. A similar trial in patients with metastatic HER2-negative breast cancer demonstrated a clinical benefit rate of 68.8% for vantictumab combined with paclitaxel; however, bone toxicity and fragility fractures limited the potential for further development [405]. The Fab F2.A is another antibody modality specific for FZD receptors; binding FZDs 1, 2, 4, 5, 7, and 8 to inhibit Wnt binding [406]. In RNF43-mutant PDAC cells, which rely on Wnt signaling for proliferation, F2.A reduced expression of Wnt target genes NKD1 and AXIN2 and induced G0/G1 cell cycle arrest more effectively than vantictumab [406]. In a similar study, full length IgG human antibodies specific for FZD5, IgG-2919, reduced subcutaneous and orthotopic RNF43-mutant HPAF-II PDAC xenograft growth by up to 74% in immunodeficient mice compared to control [407]. Moreover, IgG-2919 demonstrated more effective tumor control than vantictumab. OTSA101 is a radiolabeled antibody against FZD10 [408]. Indeed, OTSA101 radiolabeling with yttrium-90 (90Y-OTSA101), astatine-211 (211At-OTSA101), actinium-225 (225Ac-OTSA101), or indium-111 (111In-OTSA101) has been tested in several preclinical and clinical studies for synovial sarcoma [408,409,410,411]. In synovial sarcoma SYO-1 xenograft models, 211At-OTSA101, 225Ac-OTSA-101, and 90Y-OTSA101 accumulated in the tumor, significantly suppressed tumor growth following one injection, and prolonged survival [408,409]. However, in a phase I clinical trial for patients with synovial sarcoma (NCT01469975), the best recorded response for 90Y-OTSA101 was stable disease, with no objective responses observed, and the study was terminated due to slow enrollment [411]. A similar phase I trial (NCT04176016) was halted due to difficulty obtaining raw materials for 90Y-OTSA101. Ipafricept (OMP-54F28) is a fusion protein that combines the extracellular ligand-binding domain of FZD8 with an IgG1 Fc domain to inhibit Wnt signaling [412]. In pancreatic and ovarian cancer models, ipafricept combined with paclitaxel decreased tumor growth [413]. Furthermore, ipafricept with sequential paclitaxel reduced tumorigenicity of ovarian OMP-OV40 cells by sevenfold, suggesting depletion of the tumor-initiating CSC population [413]. Multiple phase I trials for ipafricept have been completed for patients with solid, ovarian, or pancreatic tumors [414,415,416]. Ipafricept monotherapy in solid tumors resulted in a best response of stable disease in 26.9% of patients (NCT01608867) [414]. In a subsequent trial for ipafricept in combination with carboplatin and paclitaxel, the overall response rate was 75.7%, with 94.6% of patients receiving clinical benefit (NCT02092363) [415]. Similar results were observed in a trial for pancreatic cancer (NCT02050178), where combination ipafricept with gemcitabine and nab-paclitaxel resulted in a clinical benefit rate of 81%, with 34.6% of patients achieving partial response [416]. The lack of control arm in both studies limits the ability to analyze whether the benefit was derived from ipafricept. Moreover, ipafricept development was halted due to bone toxicities, which presented barriers for commercial viability. Other groups have investigated anti-Wnt1 or R-spondin 3 (RSPO3) antibodies to target the Wnt/β-catenin pathway in cancer. Anti-Wnt1 is a monoclonal antibody specific for Wnt1 that inhibits H460 NSCLC tumor growth in mice [417]. R-spondin ligands like RSPO3 bind to leucine-rich repeat-containing GPCR (Lgr) 4/5/6 to potentiate Wnt signaling [393]. Anti-RSPO3 antibody, rosmantuzumab (OMP-131R10), has been shown to reduce self-renewal capabilities in PDX AML models and decrease the percentage of CSCs; however, clinical trials in advanced colorectal cancer (NCT02482441) were halted due to a lack of clinical efficacy [418,419].

10.2. Pharmacologic Inhibition

Numerous small molecule inhibitors of the Wnt/β-catenin pathway have also been developed, with many targeting PORCN. LKG974 (Wnt974) is a small molecule PORCN inhibitor that prevents Wnt activation [420]. In HNSCC xenograft and mouse mammary tumor virus (MMTV)-driven Wnt1 and Wnt3 tumor models, daily injections of LGK974 led to significant tumor regression [420]. LGK974 had an IC50 of 0.3 nM against Wnt-induced gene expression of AXIN2. Remarkably, LGK974 nearly completely abolished established intestinal RSPO-fusion tumors in mice in just one week of treatment [421]. Interestingly, LGK974 combined with aspirin more effectively inhibited tumorsphere formation in SW480 and SW742 CRC cell lines compared to oxaliplatin [422]. The results for several phase I clinical trials for LGK974 have recently been published. While LGK974 monotherapy for patients with advanced solid tumors (NCT01351103) did not result in any complete or partial responses, LGK974 in combination with encorafenib and cetuximab in patients with metastatic CRC (NCT0227813) produced marginally better results, with an overall response rate of 10% and disease control rate of 85% [423,424]. Bone toxicities and fractures were a major side effect in both studies. A phase II trial evaluating LGK974 in metastatic HNSCC (NCT02649530) was terminated; but whether this was related to the adverse effects observed in both phase I trials is unknown. ETC-159 is an orally bioavailable PORCN inhibitor that displays an IC50 value of 2.9nM in β-catenin reporter assays [425]. In colon cancer xenografts with RSPO fusion genes and RNF43 mutated pancreatic and ovarian xenografts, ETC-159 markedly reduced tumor growth and prevented tumor recurrence [425]. Histological analysis of the tumors revealed signs of mucinous differentiation with loss of adenocarcinoma. Phase I trials with ETC-159 in advanced solid tumors produced disappointing results, with only 2 out of 16 patients achieving stable disease as best outcome (NCT02521844) [426]. In the subsequent phase Ib trial, ETC-159 was tested in combination with pembrolizumab with slightly improved results (1 partial response, 5 stable disease). All patients have since discontinued, the majority of which due to disease progression, but the study is still considered active (NCT02521844) [427]. C59 is another PORCN inhibitor with a reported IC50 of 74 pmol/L in β-catenin STF reporter assays [428]. In nasopharyngeal carcinoma SUNEI1 and HNE1 xenograft models, C59 significantly reduced tumor growth, completely eliminating HNE1 tumors [429]. Interestingly, C59 also inhibited tumorsphere formation in HNE1 stem-like cells, suggesting that C59 can specifically inhibit stemness in HNE1 cell lines. RXC004 is a PORCN small molecule inhibitor that has progressed to phase II trials. In preclinical models, RXC004 reduced tumor growth and increased differentiation in Wnt-dependent RNF43-mutant pancreatic AsPC1 and HPAF-II and RSPO3-fusion SNU-1411 colorectal xenografts [430]. Interestingly, RXC004 synergized with anti-PD-1 to increase CD8+ T cell infiltration in CRC CT26 tumors. Moreover, RXC004 has been shown to increase survival and reduce tumor growth in RSPO-fusion SNU-1411 colorectal xenograft models when used in triplet combination with 5FU, irinotecan and oxaliplatin or doublet combination with 5FU and irinotecan [431]. One phase I study with RXC004 monotherapy in advanced solid cancers demonstrated modest results, with 5 out of 25 patients experiencing stable disease as best response (NCT03447470) [432]. Two phase II studies with RXC004 have commenced, one for patients with RNF3-mutant PDAC or biliary tract cancer that was recently completed (NCT04907851) and a second to assess the efficacy of RXC004 in combination with nivolumab in patients with RSPO or RNF43 mutant colorectal cancer (NCT04907539). The latter is actively recruiting. Other small molecule PORCN inhibitors include XNW7201 and CGX1321. XNW7201 is currently in a phase I trial for patients with advanced solid tumors (NCT03901950) and the results have not yet been released. Preliminary results from the phase I trial (NCT02675946) of CGX1321 in combination with pembrolizumab showed promising results for patients with GI tumors bearing RSPO or RNF43 mutations, with 71% of patients achieving stable disease [433].
Other small molecule inhibitors have focused on targeting β-catenin. Tegavivint (BC2059, tegatrabetan) inhibits β-catenin binding to transducing β-like (TBL1), thus preventing β-catenin transcriptional activity [434]. In a panel of desmoid tumor cell lines, tegavivint displayed IC50 values for cell proliferation and viability ranging from 47.79 to 284.7 nM [435]. In an ex vivo model of desmoid tumors, merely 100 nM of tegavivint significantly reduced cell viability. Moreover, tegavivint treatment in in vivo models of AML and multiple myeloma resulted in prolonged survival and decreased tumor burden [434,436]. Interestingly, tegavivint was found to synergize with proteasome inhibitor bortezomib, as well as HDAC inhibitor panobinostat [434,436]. The preliminary results for a phase I clinical trial (NCT03459469) with tegavivint in unresectable desmoid tumors were released in 2022, demonstrating an objective response rate of 25% and a nine-month PFS rate of 79% [437]. There is currently a phase I/II trial for tegavivint recruiting patients with recurrent lymphomas and desmoid tumors (NCT04851119). FH535 is another small molecule β-catenin inhibitor that prevents β-catenin interaction with TCF to inhibit β-catenin-mediated transcriptional activation [438]. In vitro analysis of FH535 revealed that it can potently inhibit TNBC MDA-MB-231 and HCC38 cell growth when cultured in three-dimensional (3D) collagen gels [439]. Moreover, FH535 attenuated the migratory and invasive capacity of both MDA-MB-231 and HCC38 cells at 100 nM concentrations. A later study revealed that FH535 significantly suppressed tumorsphere formation in CD24/CD44+ breast CSC [400]. FH535 has also demonstrated anti-CSC activity in pancreatic cancer models, reducing clonogenicity and CD24/CD44+ marker expression in vitro, as well as inhibiting metastasis and tumor growth in xenograft models [440,441]. Similarly, FH535 repressed expression of CD24, CD44, and CD133 CSC markers in colon cancer HT29 cells [442]. Interestingly, doxorubicin has also been identified as an inhibitor of β-catenin activity and signaling by preventing the β-catenin/Akt interaction [443]. Low-dose doxorubicin specifically reduced leukemia stem cell (LSC) populations without impacting hematopoietic stem and progenitor cells. Indeed, combining chemotherapy with low-dose doxorubicin significantly improved survival and reduced LSC numbers in the bone marrow of leukemic mice. Moreover, bone marrow transplants from the doxorubicin and chemotherapy treated leukemic mice displayed significantly reduced tumorigenicity when implanted into secondary recipients, further supporting the notion that low-dose doxorubicin directly targets LSCs [443]. FOG-001 is a first-in-class direct β-catenin inhibitor that blocks β-catenin interaction with TCF and is currently recruiting for phase I trials in advanced metastatic tumors (NCT05919264).
Several papers have also been published using FZD, DVL, and β-catenin transcription co-activator cAMP-responsive element binding (CREB)-binding protein (CBP) inhibitors to target Wnt/β-catenin. Carbamazepine is a well-known antiepileptic drug that was recently found to bind to FZD8 to inhibit Wnt signaling; however, it has yet to be fully explored as an anti-cancer agent [444]. FJ9 is a small molecule inhibitor that disrupts FZD7 binding to DVL, thus repressing Wnt-mediated signaling [445]. In NSCLC H460 and H1703 cell lines, FJ9 caused significant apoptosis. Moreover, in NSCLC H460 xenograft models, FJ9 treatment for 16 days reduced tumor volume [445]. Selective FZD7 peptide inhibitors have also been developed, with dFz7-21 having a reported IC50 of 100 nM against Wnt/β-catenin signaling [446]. While dFz7-21 reduced the LRG5-GFP+ intestinal stem cell population in organoids, it has not yet been tested as an anti-cancer agent [446]. Interestingly, niclosamide, which is an antihelminth compound, was found to specifically downregulate Dvl2 expression, resulting in antiproliferative effects in CRC HCT116 cells and suppression of tumor growth in HCT116 xenograft models [447]. Niclosamide nanosuspension (nano-NI) has improved the anti-cancer profile of niclosamide, showing improved inhibitory effects on ovarian tumorsphere formation and significantly reducing ovarian CP70 and SKOV3 tumor growth in mice [448]. One phase I trial (NCT02532114) for niclosamide in combination with enzalutamide for castration-resistant prostate cancer revealed a lack of clinical benefit, and the trial was terminated [449]. A phase II trial (NCT02519582) investigating the efficacy of niclosamide in CRC commenced in 2015; however, its status is currently unknown. Screening of a synthetic chemical library unveiled an Axin-stabilizing small molecule, IWR-3, that induces β-catenin destruction. Indeed, IWR-3 was shown to inhibit Wnt/β-catenin signaling in DLD-1 CRC and DU145 prostate cancer cells; however, its efficacy as an anti-tumor agent has yet to be investigated [450]. PRI-724 is a first-in-class CBP inhibitor that has progressed to clinical trials in PDAC (NCT01764477), advanced solid tumors (NCT01302405); however, the best response in both trials was stable disease [451,452].
To conclude, the Wnt/β-catenin signaling pathway is a fundamental regulatory mechanism and is especially relevant in the maintenance of CSCs. Targeting components of the Wnt/β-catenin pathway, such as receptors, ligands, and downstream effectors, has emerged as a promising strategy for cancer therapy. Despite challenges such as bone toxicities and limited clinical efficacy observed in some trials, the ongoing research and clinical investigations continue to explore the therapeutic potential of targeting the Wnt/β-catenin pathway in cancer treatment. Further understanding of the intricate regulation of this pathway and the development of novel targeted therapies hold the potential to significantly impact the management of various cancers in the future.

11. SOX2

SOX2 is a member of the SRY-related high mobility group (HMG) box family of transcription factors and is essential in embryonic development. SOX2 is a master transcription factor in the early embryo and is required for the regulation of other key developmental genes like ZFP148 and KIT. Deletion of the SOX2 gene is embryonic lethal SOX2 [453,454]. Functional SOX2 is required to maintain the pluripotential stemness required for further differentiation and growth of extraembryonic ectoderm tissues from the epiblast [455]. Furthermore, cells lacking SOX2 lose their pluripotential identity and differentiate into another cell type. In their seminal paper, Takahashi and Yamanaka identified that SOX2 is a key gene that when overexpressed in somatic differentiated fibroblast cells will induce a stem cell phenotype [456]. SOX2 plays key roles in the normal physiology of adult tissues. Arnold et al. determined that SOX2 is a bona fide stem cell marker in adult epithelial tissues, and SOX2+ cells are capable of self-renewal and give rise to differentiated progeny [457]. SOX2 is expressed in stem-cell-niches of adult tissues including the brain, lens, glandular compartment of the stomach, and lung [453].
The embryonic stem-cell-like gene signature is associated with more aggressive and poorly differentiated tumors, including breast, lung, skin, esophageal, glioblastoma, and bladder cancer [453,455,458]. SOX2 is a key driver of CSC-related protumor functions in many cancer types [459,460]. The overexpression of SOX2 resulted in increased proliferation, EMT signature, migration, and invasion of lung cancer cells [461]. In breast cancer, the patients whose tumors were enriched with embryonic stem cell genes, like SOX2, Nanog, and OCT4, had significantly worse overall survival compared to those with a non-stem-cell genotype [453]. In nasopharyngeal carcinoma CSCs, SOX2 expression was associated with high risk of tumor progression and metastasis [462]. SOX2 expression is significantly higher in squamous cell carcinoma (SCC) CD34+ tumor initiating cells compared to CD34 cells [463]. In colorectal cancer, SOX2 was shown to promote stemness, chemoresistance, and EMT [464]. Thus, the role of SOX2 as an integral stemness-promoting factor in CSCs has made it an attractive target for anti-cancer therapies (summarized in Table 1).

11.1. ZF-ATFs and PIPs

SOX2 lacks a binding pocket, so the development of direct SOX2 inhibitors is not yet available. Some groups have attempted to inhibit SOX2 at the transcriptional level. Stolzenburg et al. used a zinc finger artificial transcription factor (ZF-ATF) delivered by retrovirus to selectively suppress SOX2 in breast cancer cell lines [465]. The ATFs bind target gene promoter regions on the DNA, silencing the gene. Induction of the SOX2-specific ZF-ATFs, ZF-522SKD or ZF-598SKD, in breast MCF-7 and MDA-MB-435 cells resulted in decreased SOX2 mRNA and protein expression, as well as decreased cell viability and colony formation in vitro. Moreover, ZF-598SKD significantly reduced tumor growth in MCF-7 xenograft models [465]. ATF/SOX2 is another ZF-ATF against SOX2, which has been tested in mouse models of lung squamous cell carcinoma [466]. EBC2 lung cancer cells transfected with ATF/SOX2 failed to grow tumors in nude mice [466]. Other groups have used DNA-binding inhibitors to prevent SOX2 transcriptional activation [467]. Though it has not yet been tested as an anti-cancer agent, PIP-S2 successfully inhibited SOX2-DNA binding by targeting the SOX2 DNA-binding sequence, thus triggering the differentiation of pluripotent stem cells [467]. While the therapeutic translatability of this method is unclear, PIP-S2 appears to abrogate SOX2-mediated stemness. Collectively, these results demonstrate a template for upstream targeting of SOX2; however, further study is required to confirm their validity.

11.2. Peptides

Direct peptide antagonists for SOX2 have also been developed. Indeed, a SOX2-targeting peptide, peptide 42 (sP42), significantly reduced esophageal squamous cell carcinoma (ESCC) KYSE450 and TE-1 proliferation, as well as migration and invasion [458]. Furthermore, treating xenograft models of KYSE450 ESCC cells with sP42 resulted in significant tumor growth inhibition in mice and decreased metastatic foci in zebrafish [458]. Interestingly, sP42 also promoted terminal differentiation in KYSE450 tumors. More recently, a SOX2-interference peptide (SOX2-iPEP) consisting of a truncated 24 amino acid sequence from the C-terminus of SOX2, which includes a potential nuclear localization sequence and binding sites for SOX2 interactions with DNA and transcription factors, led to increased apoptotic death in MCF-7 and PA-1 cells [468].

11.3. Immunomodulatory Peptides and Vaccines

Other groups have investigated immunomodulatory peptides against SOX2. Amino-acid sequence screening of SOX2 revealed three alleles predicted to bind to human leukocyte antigen (HLA)-A*0201 to promote CD8+ T cell activation [469]. CD8+ T cells stimulated with SOX2 peptide 60030 efficiently lysed SOX2-expressing glioma cell lines. In a separate study, oligodendroglioma tumor-bearing mice “vaccinated” with SOX2 peptides in combination with temozolomide lived twice as long as both untreated mice and mice treated with temozolomide [470]. Recently, several phase I and II clinical trials (NCT05242965, NCT05455658, NCT02157051) have commenced for STEMVAC, a DNA plasmid-based vaccine that targets multiple cancer antigens, including SOX2. These studies are currently ongoing, and the results have not yet been published.

11.4. Pharmacologic Inhibition

The majority of pharmacologic inhibitors targeting SOX2-dependent cancer growth do not target SOX2 directly but inhibit SOX2-associated signaling pathways and molecules. SOX2 is involved in many cancer pathways, and the PI3K/Akt/mTOR pathway has been shown to positively regulate SOX2 expression [471]. Rapamycin is a specific mTOR complex 1 inhibitor that recently gained FDA approval for advanced perivascular epithelioid cell tumors [472]. Interestingly, rapamycin decreased SOX2 mRNA expression in glioma U251 and U373 cell lines and inhibited U251 glioma tumor growth in mice [473]. Other groups have focused on targeting Akt, as Akt has been shown to support SOX2 overexpression by preventing its ubiquitination and degradation by the proteosome in ESCC [474]. Indeed, a study of ESCC determined that SOX2 levels do not correlate directly to gene copy number but instead to cellular Akt levels [474]. Indeed, MK2206, which is an Akt inhibitor, reduced SOX2 protein levels and suppressed tumor spheroid formation in ESCC K450 cells [474]. MK2206 has been investigated in a phase II trial (NCT01277757) for patients with advanced breast cancer; however, it displayed very little clinical efficacy [475]. Similarly, Qin et al. showed that Akt inhibitor DC120 was able to indirectly downregulate SOX2 protein and mRNA levels in nasopharyngeal carcinoma CSCs by upregulating tumor suppressor p27 [476]. In a preclinical model of nasopharyngeal carcinoma, DC120 significantly reduced tumor volume and CSC in both primary and secondary xenograft tumors, which they attributed to a decrease in SOX2 expression [476].
SOX2 is also a downstream target of growth receptor pathways. Indeed, SOX2 expression is stimulated by fibroblast growth factor receptor 1 (FGFR1) activation in lung cancer [461]. FGFR1-amplification in lung cancer cell lines H1581 and DMS1144 was shown to promote EMT, migration, and invasion, which were attributed to SOX2 expression [461]. Mechanistically, FGFR1 promotes ERK1/2 phosphorylation, which promotes the expression of SOX2. AZD4547 is a FGFR1 inhibitor that markedly suppressed SOX2 expression and tumor growth in orthotopic and subcutaneous H1581 and DMS1144 lung cancer xenograft models [461]. More recently, AZD4547 has been investigated in phase II clinical trials (NCT01791985) for endocrine-resistant breast cancer in combination with anastrozole or letrozole; however, only 10% of patients exhibited partial response [477]. SOX2 expression is also modulated by epidermal growth factor receptor (EGFR)/Src/Akt signaling pathways. Indeed, inhibition of EFGR/Src/Akt signaling with EFGR inhibitor gefitinib, Src inhibitor dasatinib, and Akt inhibitor LY294002 specifically decreased SOX2 expression in NSCLC H1650SP-Adh CSCs [478]. Moreover, EFGR inhibitors gefitinib and erlotinib both decreased the proportions of CSCs in NSCLC and reduced spheroid formation and size [478]. Currently, gefitinib and erlotinib, as well as dasatinib, are FDA-approved for treatment of NSCLC and some forms of leukemia, respectively. Interestingly, gentian violet, which is an antimycotic and antibacterial agent, was found to reduce EFGR activation and STAT2 phosphorylation, thus preventing STAT2 binding to the SOX2 promoter [479]. Indeed, gentian violet treatment impaired melanoma CSCs by inhibiting SOX2 expression, resulting in reduced sphere-forming capacity, with enhanced effects seen on second generation spheroids, in a dose-dependent manner. The effect was abolished with SOX2-overexpression [479]. Together, this is indicative that growth factor receptor inhibitors decrease stem cell capabilities in multiple SOX2-driven cancer types.
To identify other indirect targets of SOX2, Kim et al. studied the interactome and found that SOX2 interacts with transcription factors, chromatin remodelers and modifiers, cyclin-dependent kinases, and others [480]. They identified EP300 as a key mediator of SOX2 expression, and CBP30, which is an EP300 inhibitor, reduced MGH7 and ChaGoK1 lung cancer cell growth in vitro [480]. Interestingly, CBP130 synergized with PI3K inhibitor, BKM120, to further decrease growth [480]. Others have identified pevonedistat (MLN4924), a neddylation inhibitor, as a potent indirect SOX2 inhibitor [481]. Mechanistically, pevonedistat inactivates the E3 ligase F-Box and WD repeat domain containing 2 (FBXW2), preventing Msh homeobox 2 (MSX2) ubiquitylation and degradation. MSX2 acts as a SOX2 transcription repressor, and pevonedistat-mediated MSX2 stabilization results in decreased SOX2 mRNA and protein expression [481]. Pevonedistat sensitized the tamoxifen-resistant MCF-7 cells, resulting in significant tumor growth control. Pevonedistat in combination with various chemotherapy agents has been tested in several clinical trials (NCT03268954, NCT03323034, NCT03770260, NCT03965689), with four patients achieving complete response in a trial for AML [482,483]. Identifying the coactivators of SOX2 may be an effective method of targeting the tumorigenic properties it bestows.
Other groups have targeted factors impacting SOX2 protein stability. In a preclinical model of nasopharyngeal carcinoma, XIAP maintained stemness by stabilizing the SOX2 protein, extending its half-life by inhibiting autophagic degradation [462]. The drug APG-1387 is a second mitochondria-derived activator of caspases (SMAC) mimetic that directly binds to X chromosome-linked inhibitor of apoptosis protein (XIAP), leading to the induction of apoptosis [484]. APG-1387 has shown anti-CSC activity, potently reducing CSC populations in nasopharyngeal carcinoma S-18 cells [462]. In primary S-18 xenograft mouse models, APG-1387 reduced tumor growth compared to control and synergized with 5FU or cisplatin, resulting in a significant reduction in tumor growth. Moreover, secondary xenografts displayed significantly slower growth following treatment with APG-1387 alone or in combination with 5-FU or cisplatin, indicating at least a partial eradication of CSCs.
The targeting of epigenetic mechanisms to repress SOX2 has also been investigated as an anti-cancer mechanism. Histone deacetylase 11 (HDAC11) is a chromatin remodeler that has recently been implicated as an inducer of SOX2 expression in NSCLC [485]. Selective HDAC11 inhibitors, FT234 and FT895, reduced SOX2 expression in H1650 and A549 NSCLC cells and suppressed stemness properties, evidenced by reduced self-renewal and tumorsphere formation [485]. Similarly, lysine-specific demethylase 1 (LSD1) demethylates lysine residues on histone H3 and is an essential epigenetic regulator of pluripotency [486,487]. Moreover, LSD1 is required for SOX2 expression in lung cancer H520, A2780, and T47D cells and is positively correlated with SOX2 expression in lung squamous cell carcinoma patient samples [486]. Use of an LSD1 inhibitor, CBB1007, enhanced repressive histone H3 methylation at the SOX2 enhancer, resulting in the reduced binding of SOX2 to its target genes [486]. Iadademstat (ORY-1001) is a highly potent LSD1 inhibitor that suppressed enhancer-driven activation of SOX2 in BT-474 breast cancer cells [487]. Moreover, iadademstat caused a dose-dependent decrease in tumor spheroid formation in MDA-MB-436 TNBC cells with no effect on viability assays, demonstrating that the drug effectively and specifically targets stemness potential without off-target effects [487]. Phase I trials for iadademstat have been completed in AML (EUDRACT 2013-002447-29), with an objective response rate of 36% [488]. Other clinical trials for iadademstat in combination with paclitaxel in SCLC or gilteritinib in AML are currently recruiting (NCT05420636, NCT05546580).
SOX2 is a pivotal player in both normal physiological processes and cancer progression, particularly as a driver of CSC properties across various cancer types. It has been clearly demonstrated that targeting SOX2 is a potent method of inhibiting CSCs and their tumorigenic potential. While direct inhibition of SOX2 has posed challenges due to its lack of a binding pocket, alternative strategies targeting its transcriptional regulation, protein stability, and epigenetic modulation have shown promise in preclinical and clinical studies. Further research and clinical trials are warranted to validate and optimize these approaches for clinical use in cancer therapy.

12. Hedgehog

The hedgehog (HH) pathway is a signal transduction pathway that confers gene expression changes, activated by HH ligands. The pathway was first discovered in 1980 in drosophila melanogaster and determines segment polarity [489]. There are three mammalian HH pathway ligands sonic hedgehog (SHH), Indian hedgehog (IHH), and Desert Hedgehog (DHH). SHH and IHH are present in most tissues, and SHH is the most extensively studied [490]. DHH is present in gonads [491]. HH signaling is essential in determining the cell fate, cell patterns and signals between cell growth and differentiation [490]. The canonical HH pathway involves two transmembrane receptors smoothened (SMO), a G-protein coupled receptor and patched 1 (PTCH). Upon ligand binding to PTCH, a SMO repressor, SMO is accumulated in primary cilium and downstream processing and activation of glioma-associated oncogene (GLI) family transcription factors are initiated [492]. Further, GLI proteins activate transcription of its targets. The HH pathway is silenced in adult tissue unless aberrant activation in case of certain cancers. Several cancers have aberrant HH pathway activation. It is well appreciated that HH signaling is important for the maintenance of CSCs across multiple cancer types [489,490]. Indeed, HH signaling components are overexpressed in CD24/CD44+ breast CSCs [491]. Similarly, HH signaling was found to be essential for maintenance of CSCs in chronic myelogenous leukemia (CML), as SMO−/− cells displayed reduced tumorigenicity in CML mouse models [493]. Isolated ALDH+ melanoma CSCs display enhanced tumorigenicity and self-renewal capabilities, effects which are abrogated upon HH signaling inhibition [492]. In pancreatic cancer, HH signaling was found to regulate pancreatic CD133+ CSC properties such as tumorsphere formation [494]. Thus, targeting the components of HH signaling present attractive anti-cancer targets (Figure 4, Table 1).

12.1. Pharmacologic Inhibitors

With respect to inhibition of the HH pathway, most drugs inhibit the HH target SMO, with several receiving FDA approval as anti-cancer agents [495]. Vismodegib (GDC-0449) is a small molecule antagonist of SMO and was the first FDA approved HH pathway inhibitor for cancer [496]. In preclinical studies on pancreatic cancer CSCs, vismodegib suppressed primary and secondary spheroid formation, indicating that it can directly inhibit CSC characteristics [497]. Keyword searches in the ClinicalTrials.gov database showed 85 different studies in several solid tumors studying vismodegib as a monotherapy or in combination, for which results have been extensively reviewed elsewhere [496]. Vismodegib monotherapy in phase I and II trials showed promise in basal cell carcinoma (BCC) patients with high overall response rates (NCT00607724, NCT00833417) [498,499]. However, clinical trials in other cancer types including refractory gastric cancer (NCT03052478) and ovarian cancer (NCT00739661) showed less promising results [500,501]. In a phase II clinical trial for metastatic PDAC (NCT01088815), vismodegib in combination with gemcitabine and paclitaxel showed better response with compared to monotherapy, with a 40% objective response rate [502]. Sonidegib (LDE225) is the second drug following vismodegib to be approved by FDA for BCC in 2015 [503]. Irvine et al., 2016 showed sonidegib treatment by itself or in combination with nilotinib (tyrosine kinase inhibitor against BCR-ABL oncogene of CML) reduced the self-renewal capacity of chronic phase (CP) CML in vitro and when engrafted to NSG mice [504]. Further, sonidegib reduces multi-drug resistance p glycoproteins ABCB1 and ABCG2, while their gene levels are not altered in NSCLC, this increases cytotoxicity of other drugs like mitotraxane and daunorubicin [505]. Clinical trials using sonidegib have shown high success rates in some cancers [503]. Indeed, in BCC, sonidegib monotherapy showed 56% ORR (NCT01327053) while combination therapy using etoposide and cisplatin on extensive SCLC showed 79% partial response (NCT01579929) [506,507]. In a small phase I trial, sonidegib in combination with docetaxel led to complete response in one patient (NCT02027376) [508]. Sonidegib has also been tested in clinical trials for medulloblastoma (NCT01125800), and CML (NCT01456676) with varying degrees of efficacy [503]. Glasdegib (PF-04449913) is another SMO inhibitor with FDA-approval that has been used in clinical trials, mainly for treating acute myeloid leukemia [509,510]. In the phase II BRIGHT AML 1003 clinical trial (NCT01546038), glasdegib improved the efficacy of low dose cytarabine (LDAC) treatment in myeloid leukaemia patients ineligible for intensive chemotherapy, improving complete remission from 2.6% in LDAC treated patients to 19.2% patients in combination therapy [511]. However, in the phase 3 BRIGHT AML 1019 study (NCT03416179) on patients with untreated AML, glasdegib showed no benefit with intensive treatment of cytarabine or danorubicin, and the efficacy endpoints of the study were not met [512].
Cyclopamine is a steroidal alkaloid that directly binds to the heptahelical bundle of SMO, however it displayed teratogenic properties which limited its clinical translatability [513,514]. Second generation derivatives of cyclopamine have yielded several HH-inhibiting compounds including vismodegib (described above), IPI-269609, and saridegib (IPI-926). IPI-269609 reduced Gli1 mRNA, as well as the ALDH+ pancreatic E3LZ10.7 cell population, and impaired colony formation [515]. Later modifications of IPI-269609 yielded saridegib, a potent SMO inhibitor which completely abrogated tumor regrowth in B837Tx medulloblastoma xenograft models following treatment with saridegib, indicating complete elimination of cancer cells [516]. In chondrosarcoma xenograft tumors, saridegib inhibited GLI1 expression and significantly reduced tumor volume in mice [517]. Moreover, histological analysis revealed increased calcification in the tumors, suggesting that IPI-926 may drive chondrosarcoma differentiation [517]. Multiple clinical trials have been completed for saridegib with relatively modest results [495]. In a small pilot study for HNSCC (NCT01255800), saridegib in combination with cetuximab yielded one partial response, and a median PFS of 77 days [518]. IPI-926 showed some anti-tumor activity in a separate phase I clinical trial for HNSCC, with two complete responses and six partial responses in patients with BCC [519]. FOLFIRINOX (5FU, leucovirin, irinotecan, oxaliplatin) in combination with IPI-926 displayed an unconfirmed objective response rate of 66.7% in patients with pancreatic cancer (NCT01383538) [520]. A double-blind placebo-controlled phase II trial for chondrosarcoma, revealed that IPI-926 therapy had no effect on PFS, or overall survival compared to placebo (NCT01310816) [521].
Other SMO inhibitors include BMS-833923 (XL139), taladegib (LY2940680, ENV-101), and Hh003. BMS-833923 was tested in esophageal adenocarcinoma cell lines and was shown to decreased cell proliferation and increase apoptosis [522]. In ovarian cancer cell lines, BMS-833923 synergized with carboplatin to induce cell death [523]. Moreover, BMS-833923 was shown to significantly decrease the quiescent CD34+/CD38 LSC population in AML in vitro [524]. BMS-833923 plus dasatinib has also been investigated in a phase I clinical trial (NCT01218477) for patients with CML; however, it displayed very limited clinical efficacy [525]. Taladegib was shown to potently inhibit HH signaling in medulloblastoma cell lines, and significantly prolonged survival, and inhibited tumor growth of Ptch+/− p53−/− transgenic mice which spontaneously develop medulloblastoma [526]. Multiple clinical trials for taladegib have been completed, (NCT01919398, NCT02784795). In one phase I trial, taladegib displayed a 46.8% response rate in patients with BCC, five of which were complete response (NCT01226485) [527]. There is currently one active phase II study evaluating taladegib advanced solid tumors with PTCH1 loss of function mutations (NCT05199584). While Hh003 has not yet been tested in clinical trials, it was shown to inhibit the colony formation of colorectal HCT116 and pancreatic Panc-1 cells more effectively than vismodegib [528]. Furthermore, Hh003 significantly suppressed HCT116 tumor growth in mice compared to control and vismodegib treatment [528].
Itraconazole is an azole-based FDA-approved antifungal drug, which has been repurposed in preclinical and clinical trials as an anti-cancer agent [529]. The exact mechanism by which itraconazole inhibits HH signaling is not entirely clear, with some groups suggesting SMO inhibition, and others implicating inhibition of Gli1 and HH expression [530,531]. Itraconazole treatment in mouse xenograft models of medulloblastoma, colorectal, and breast cancer impaired tumor growth [530,531,532]. In all these studies inhibition of HH signaling and reduction of GLI levels were observed. Retrospective analysis of patients with TNBC, metastatic pancreatic cancer or biliary tract cancer who received chemotherapy followed by itraconazole revealed promising results, with median overall rates ranging 47–62% [533,534,535]. In a phase II clinical trial (NCT00769600) investigating itraconazole in combination with pemetrexed in NSCLC, itraconazole improved significantly PFS compared to pemetrexed alone, with overall survival increasing from 8 months to 32 months [536]. High dose itraconazole also increased prostate-specific antigen progression-free survival rates to 48% at 24 weeks in a phase II clinical trial for castration resistant prostate cancer [537].
Arsenic based drugs (arsenicals) inhibit GLI2, activator of HH dependent transcription [495]. Three different arsenicals, Sodium arsenite, Arsenic trioxide (ATO) and phenylarsine oxide showed reduction of HH signalling in medulloblastoma cells, resulting in their death [538]. ATO binds to GLI family transcription factors to inhibit their ciliary localization and further nuclear translocation in medulloblastoma and in promyelocytic leukemia [538,539]. In SCLC CSCs, ATO significantly inhibited GLI1 expression, resulting in reduced clonogenicity and spheroid formation, as well as tumorigenicity SCLC xenograft models [540]. Similar effects were observed when Ewing Sarcoma cells were treated with ATO, reducing GLI1 levels, resulting in decreased tumor growth [541]. Moreover, in pancreatic cancer xenografts, ATO in combination with gemcitabine reduced CD24/CD44+ and ALDH1A1+ CSC populations and inhibited tumor growth [542].
GANT61 is a small molecule which inhibits GLI1/2 DNA binding [543]. GANT61 increased apoptosis in biliary tract cancer cell lines in vitro [544]. In melanoma tumorspheres with CSC characteristics and high ALDH expression, GANT61 significantly reduced tumorsphere formation, suggesting targeting of the CSC population [492]. Similarly, GANT61 abolished tumorsphere formation in CD133+ pancreatic cancer cells and undifferentiated HLE and HLF hepatocellular carcinoma cells, further implicating GANT61 as a direct CSC inhibitor [494,545]. GANT61 has not yet progressed to clinical trials, likely due to relatively high doses required to see anti-CSC effects.

12.2. Antibodies

Only a few antibodies targeting the HH signaling pathway have been explored for anti-cancer activity. MEDI-5304 is a human antibody with specificity for both SHH and IHH [546]. Interestingly, despite demonstrating anti-tumor activity in HT-29 colon xenografts at a dose of 1 mg/kg, MEDI-5304 did not reduce tumor growth in pancreatic P479 subcutaneous xenografts, or the frequency of CSCs in the pancreatic tumor explants [546]. Whether MEDI-5304 reduced CSC frequency in CRC was not examined. The authors also tested the efficacy of 5E1, which is a mouse mAb specific for SHH [496]. While MEDI-5306 displayed high SHH binding affinity than 5E1, both antibodies displayed similar anti-tumor activity in CRC xenograft models [546]. In pancreatic PDX tumor models, 5E1 displayed moderate anti-cancer activity, preventing tumor growth and even facilitating tumor regression [547]. In a mouse model of breast cancer, 5E1 has demonstrated the ability to reduce both tumor size and the occurrence of liver and pancreatic metastases [548]. Similarly, 5E1 decreased primary HCT8 colon xenograft tumor size in nude mice, as well as lung metastasis in chorioallantoic membrane chick embryo models [549]. α-Ptch1 is a novel anti-PTCH1 antibody which reportedly reduced proliferation of pancreatic cancer cells; however, the data on this antibody is limited [550].
Targeting the HH signaling pathway shows promise as a therapeutic approach for various cancers; however, there are challenges that need to be addressed. Pharmacological inhibitors such as vismodegib, sonidegib, and glasdegib have demonstrated efficacy in clinical trials, but one limiting factor is the potential development of resistance due to acquired mutations in the SMO receptor. Despite this challenge, second-generation inhibitors and repurposed drugs offer alternatives for overcoming resistance and improving treatment outcomes. Ongoing research into novel inhibitors and combination therapies, along with a deeper understanding of the mechanisms underlying HH signaling, are essential for maximizing the therapeutic potential of targeting this pathway in cancer treatment. Additionally, efforts to develop antibody-based therapies targeting HH signaling components may provide new avenues for intervention in HH pathway-driven malignancies.

13. Conclusions

In this review, we have presented a comprehensive summary of the therapeutic targets associated with CSCs, specifically focusing on the CSC markers and pathways that are most prevalent across multiple cancer types. The studies on CSCs have highlighted pathways and factors that are important in tumor progression and recurrence and thus present as attractive targets, regardless of their association with CSCs. Our analysis highlights the diverse array of molecular targets associated with CSC stemness, including ALDHs, CD44, EpCAM, CXCR4, Sox2, CD55, CD133, and signaling pathways such as Wnt/β-catenin, Notch, and Sonic Hedgehog. Indeed, the therapeutic landscape for therapies that target these cancer-promoting factors that have been ascribed to CSCs is expansive and diverse. From pharmacologic small molecule inhibitors and antibody–drug conjugates to immunomodulatory agents such as antibodies, CAR-T cells, CAR NK cells, BiTEs, immunotoxins, and vaccines, there exist a plethora of strategies that could be used to target CSCs. It is important to note that although these factors are associated with CSCs, many of the studies discussed were not specifically aimed at targeting CSCs as a read out of efficacy and often assessed only if tumor growth was impeded. They may have found the unintended beneficial side-effect of reducing CSCs if this had been assessed.
Despite the remarkable progress made in developing therapies against factors and markers associated with CSCs, several challenges persist in translating these findings into clinically effective therapies. Notably, many of the molecular targets and pathways associated with CSCs are also expressed in normal physiological processes and other stem cell populations. This presents a significant hurdle, as on-target effects of therapeutic interventions could inadvertently impact normal tissue function, leading to potential toxicity and adverse effects. Furthermore, while preclinical studies have demonstrated the efficacy of these novel agents in inhibiting tumor growth and metastasis, the translation of these findings into successful clinical outcomes has been limited. While clinical studies have provided invaluable insights into the efficacy of targeting factors associated with CSC therapies, none have yet been approved as specific anti-CSC treatments. Instead, the therapeutic agents that have gained regulatory approval function as broad-spectrum anti-cancer agents, coincidentally targeting molecules and pathways that are elevated in CSCs. This underscores the complexity of CSC biology and the challenges associated with developing therapies that selectively eradicate CSCs while sparing normal cells.
Moving forward, addressing the specificity of the targeted therapies will be paramount to mitigate off-target effects and enhance therapeutic efficacy. Additionally, continued research efforts are needed to identify novel CSC-specific vulnerabilities and refine therapeutic strategies that exploit these targets. By leveraging the collective knowledge gained from preclinical investigations and clinical trials, we can advance the development of more precise and effective targeted therapies that in addition to inhibiting tumor progression, target CSCs and ultimately improve the long-term outcomes for cancer patients.
Table 1. Drugs targeting CSC markers and pathways with types of cancer tested in and clinical status.
Table 1. Drugs targeting CSC markers and pathways with types of cancer tested in and clinical status.
CSC TargetDrugConditionClinical StatusReference/Trial Number (if Applicable)
ALDH1A1NCT-501HNSCC, pancreatic, CRCPreclinical[42,43,44]
CM37OvarianPreclinical[45]
Compound 974OvarianPreclinical[46]
ALDH1A3GA11GliomaPreclinical[50]
MF-7BreastPreclinical[12]
NR6Glioblastoma, CRCPreclinical[51]
MCI-INI-3GliomaPreclinical[52]
YD1701CRCPreclinical[53]
ALDH2CVT-10216CRCPreclinical[13]
DaidzinCRCPreclinical[13]
ALDH3A1DyclonineSCC, gastricPreclinical[60]
CB57Lung, glioblastomaPreclinical[61]
CB29GlioblastomaPreclinical[62]
EN40LungPreclinical[64]
Pan-ALDHDEABMelanoma, pancreaticPreclinical[66,67,68]
NanoKS100MelanomaPreclinical[69]
DIMATEAMLPreclinical[70,71]
637AOvarianPreclinical[72]
CitralBreastPreclinical[73]
DisulfiramBreast, lungPreclinical[73,74,75]
EpCAMEpAb2-6CRC, SCC, pancreatic, lung, Preclinical[90,91]
Adecatumumab (MT201)Ovarian, breast, prostatePhase II[93,94,95]
AM-928Solid tumors Phase INCT05687682
Solitomab (MT110)Pancreatic, ovarian, solid tumors, Phase I[98,99,100]
CatumaxomabMalignant ascitesApproval withdrawn in European Union[101]
EpCAM-CD3 hFc mRNA-LNPOvarianPreclinical[102]
Anti-EpCAM CAR-T cellsOvarian, prostate, lung, gastric, pancreaticPreclinical[103,104,105,106]
IMC001GI tumorsPhase II[108]
NCT05028933, NCT04196465
VB6-845Ovarian, breast, SCLC, CRC, SCCPhase I[109,110,111] NCT00481936
VB4-845HNSCC, bladder carcinomaPhase III[113,114,115,116] NCT04859751
SyntOFFBreastPreclinical[119]
chiHEA125-AmaPancreaticPreclinical[120]
Anti-EpCAM siRNABreast, retinoblastoma Preclinical[123,124]
CD44Bivatuzumab (BIWA-4)HNSCCPhase I[133]
NCT02254018
H4C4PancreaticPreclinical[134]
IM7BreastPreclinical[135]
RG7356 (RO5429083)Leukemia, HNSCC, solid tumorsPhase I[136,137,138,139,140] NCT01358903, NCT01641250
Anti-CD44-IR700 NIR-PITSCC, CRC, lungPreclinical[142,143,144]
rhPRG4BreastPreclinical[145,146]
Apt#7BreastPreclinical[149]
CD44-EpCAM aptamerOvarianPreclinical[150]
ASO 4401HCCPreclinical[152]
THIQHNSCCPreclinical[154,155]
JE22-NPBreastPreclinical[156]
VerbascosideGlioblastomaPreclinical[157]
A6 (SPL-108)Prostate, multiple myeloma, breast, ovarian, leukemiaPhase II[158,159,160,161,162,163,164,165] NCT00939809, NCT02046928
CD55Anti-CD55CRCPreclinical[172]
MB55Lymphoma, leukemiaPreclinical[173]
CD55 NAbNeuroblastomaPreclinical[168]
177Lu-anti-CD55LungPreclinical[174]
GB262PancreaticPreclinical[175]
105AD7CRC, osteosarcomaPhase II[176,177,178,179]
PAT-SC1GastricPhase I[180]
CD55spCervicalPreclinical[181]
C-PC/CMC-CD55CervicalPreclinical[182]
AWT-489CRCPreclinical[184]
CRISPR/cas9CervicalPreclinical[171]
siRNABreast, ovarian, lungPreclinical[185]
CXCR4Ulocuplumab (BMS-936564, MDX-1338) Leukemia, lymphoma, multiple myeloma, breast, Waldenström macroglobulinemia Phase II[199,200,201,202,203] NCT01120457, NCT02472977, NCT02305563, NCT03225716, NCT01359657, NCT02666209
PF-06747143LeukemiaPreclinical[204,205,206]
12G5Endometrial, osteosarcoma Preclinical[207,208]
ALX-0651N/APhase I[209]
NCT01374503
LY2624587Lymphoma, leukemiaPhase I[210]
NCT01139788
Hz515H7
(F50067)
Multiple myeloma, lymphoma, AMLPhase I[211,213]
MEDI3185Multiple myeloma, Burkitt’s lymphoma, ovarian, lungPreclinical[212]
Plerixafor (AMD3100, Mozobil®)Breast, lung, CRC, prostate, pancreatic,FDA approved for multiple myeloma and lymphoma[214,215,216,217,218,219,220,221,222,223,224]
NCT00694590, NCT05510544, NCT00903968, NCT00906945
Mavorixafor (X4P-001)RCC, melanoma, breast, Waldenström’s macroglobulinemiaPhase II[225,226] NCT02823405, NCT05103917, NCT02667886, NCT02923531, NCT04274738
USL311GlioblastomaPhase I/IINCT02765165
PRX177561GlioblastomaPreclinical[227]
MSX-122Breast, HNSCC, uveal melanoma Phase I[228]
NCT00591682
Motixafortide (BL-8040, BKT-140, TNI4001)AML, breast, pancreatic, solid tumorsPhase II[218,228,229,230,231,232]
NCT01838395, NCT02826486
LY2510924AML, solid tumors, RCC, SCLCPhase II[233,234,235]
NCT02737072, NCT02652871, NCT01391130, NCT01439568
CTCE-9908Breast, solid tumorsPhase I/II[236,237]
IS4Prostate, melanomaPreclinical[238]
CD133Anti-CD133 mAbCRC, breastPreclinical[256,257,258]
BsAb-CIKPancreaticPreclinical[259]
293C3-SDIECRC, leukemiaPreclinical[260,261]
Anti-CD133 CAR NK cellsOvarianPreclinical[262]
Anti-CD133 CAR T cellsSCLC, liver, CRCPhase II[263,264,265]
NCT02541370, NCT02541370
16x133 BiKECRCPreclinical[266]
16x15x133 TriKECRC, breast, HNSCC, prostate, AMLPreclinical[267]
1615EpCAM133 TetraKECRCPreclinical[268]
ICT-121GlioblastomaPhase I[269]
NCT02049489
GMILungPreclinical[270]
dCD133KDELHNSCC, breast, ovarianPreclinical[271,272,273]
AC133-saporinCRCPreclinical[274]
CelecoxibCRCFDA-approved NSAID for migraines[275,276]
TrifluridineCRCFDA-approved for metastatic CRC[277]
ACT001Lung, gliomaPhase II[278,279,280]
ACTRN12616000228482, NCT05053880
CD133 aptamersHCC, breastPreclinical[281,282,284]
LS-7CRC, breastPreclinical[285]
CRISPR/cas9CRCPreclinical[286]
siRNACRCPreclinical[287]
AC133 NIR-PITGliomaPreclinical[288]
NanogIGT-PMOBreastPreclinical[302]
siRNACRCPreclinical[305]
SAHAHNSCC, lymphomaFDA-approved for T-cell lymphoma [307,308]
PiBProstatePreclinical[309]
ResveratrolGlioblastomaPreclinical[313,314]
AspirinCRCFDA-approved NSAID[315]
MetforminTNBCFDA-approved for diabetes mellitus[316]
DFX, SP10EsophagealPreclinical[317]
NotchCrenigacestat (LY3039478)Breast, CRC, lung, ovarian, glioblastoma, gastric, intrahepatic cholangiocarcinoma, multiple myelomaPhase I[327,328,329,330,331]
NCT01695005, NCT02836600,
NCT03502577
LY900009Solid tumors, lymphoma Phase I[333]
NCT01158404
Osugacestat (AL101, BMS-906024)Breast, leukemia, adenoid cystic carcinoma, NSCLCPhase II[334,335,336,337,338]
NCT03691207, NCT01653470
RO4929097 (RG473)Melanoma, CRC, sarcoma, pancreatic adenocarcinomaPhase II[340,344]
NCT01116687, NCT01120275, NCT01154452, NCT01232829
Nirogacestat (PF-03084014)HCC, prostate, dermoid, breastPhase III[345,346,347,348,349]
NCT01981551, NCT04195399, NCT03785964, NCT01876251
DAPTOsteosarcoma, gastric, adenomaPreclinical[350,351,352]
MRK-0752Breast, ovarian, solid tumors, PDAC, CNS malignanciesPhase II[326,353,354,355,356,357]
NCT01098344
MRK-560LeukemiaPreclinical[358]
Limantrafin (CB103)Breast, leukemia Preclinical[359,360]
ZLDI-8NSCLC, CRC, HCCPreclinical[361,362,363]
602.101BreastPreclinical[364]
Anti-Notch1 mAbLung, CRCPreclinical[365]
Tarextumab (OMP-59R5)Breast, lung, ovarian, pancreaticPhase II[366,367,368,369]
NCT01277146,
NCT01859741, NCT01647828
Brontictuzumab (OMP-52M51)Solid tumors, hematologic and lymphoid malignancies, CRC, ACC,Phase I[370,371]
NCT01778439,
NCT01778439
PF-06650808BreastPhase I[372]
NCT02129205
Enoticumab (REGN421)Solid tumorsPhase I[373]
NCT00871559
Demcizumab (OMP-21M18)Ovarian, peritoneal, fallopian, NSCLC, pancreatic, solid tumorsPhase I[374,375]
NCT01952249,
NCT01189968,
NCT01189942,
NCT02722954,
NCT01952249
MEDI0639Solid tumorsPhase I[376,377]
NCT01952249
MGZ01BreastPreclinical[378]
Navicixizumab (OMP-305B83)Solid tumors, ovarian, peritoneal, fallopianPhase I[379,380]
NCT02298387,
NCT03030287
ABT-165Glioblastoma, CRCPhase I[381,382]
NCT03368859
Tarlatamab (AMG 757)SCLCPhase II[383,384,385]
NCT03319940,
NCT05060016
HPN328SCLC, neuroendocrine Phase I/II[386]
NCT04471727
Rova-TSCLCPhase III[387,388]
NCT04471727
Wnt/β-cateninVantictumab (OMP-18R5)CRC, breast, lung, pancreatic, solid tumors,Phase I[402,403,404,405]
NCT01345201,
NCT02005315,
F2.APDACPreclinical[406]
IgG-2919PDACPreclinical[407]
OTSA101Synovial sarcomaPhase I[408,409,410,411]
NCT01469975,
NCT04176016
Ipafricept (OMP-54F28)Pancreatic, ovarian, solid tumorsPhase I[413,414,415,416]
NCT01608867,
NCT02092363,
NCT02050178
Rosmantuzumab (OMP-131R10)Leukemia, CRCPhase I[418,419]
NCT02482441
LGK974 (Wnt974)HNSCC, CRC, solid tumorsPhase II[420,421,422,423,424]
NCT01351103, NCT0227813,
NCT02649530
ETC-159CRC, pancreatic, solid tumorsPhase I[425,426,427]
NCT02521844,
NCT02521844
C59Nasopharyngeal carcinomaPreclinical[428,429]
RXC004Pancreatic, CRC, solid tumorsPhase II[430,431,432]
NCT03447470,
NCT04907851,
NCT04907539
XNW7201Solid tumorsPhase INCT03901950
CGX1321GI tumorsPhase I[433]
NCT02675946
Tegavivint (BC2059, tegatrabetan)Desmoid, AML, multiple myelomaPhase I/II[434,435,436,437]
NCT03459469,
NCT04851119
FH535Breast, pancreatic, CRCPreclinical[400,438,439,440,441,442]
DoxorubicinLeukemiaPreclinical[443]
FOG-001Solid tumorsPhase INCT05919264
CarbamazepineN/AFDA-approved anti-epileptic[444]
FJ9NSCLCPreclinical[445]
dFz7-21N/APreclinical[446]
NiclosamideCRC, ovarian, prostateFDA-approved anti-helminthic[447,448,449]
NCT02532114,
NCT02519582
SOX2ZF-522SKD/
ZF598SKD
Breast Preclinical[465]
ATF/SOX2LungPreclinical[466]
PIP-S2N/APreclinical[467]
sP42ESCCPreclinical[458]
SOX2-iPEPBreast, ovarianPreclinical[468]
60030OligodendrogliomaPreclinical[470]
STEMVACBreast, NSCLCPhase IINCT05242965, NCT05455658, NCT02157051
RapamycinGliomaFDA-approved for perivascular epithelioid tumors[473]
MK2206ESCC, breastPhase II[474,475]
NCT01277757
DC120Nasopharyngeal carcinomaPreclinical[476]
AZD4547LungPhase II[461,477]
NCT01791985
GefitinibNSCLCFDA-approved for NSCLC[478]
DasatinibNSCLCFDA-approved for CML[478]
LY294002NSCLCPreclinical[478]
ErlotinibNSCLCFDA-approved for NSCLC[478]
Gentian violetMelanomaFDA-approved antimycotic/antibacterial[479]
CBP30LungPreclinical[480]
Pevonedistat (MLN4924)Breast, NSCLC, leukemia, multiple myeloma, solid tumors, lymphomaPhase III[481,482,483]
NCT03268954, NCT03323034, NCT03770260, NCT03965689
APG-1387Nasopharyngeal carcinoma Preclinical[462,484]
FT234, FT895NSCLCPreclinical[485]
CBB1007LungPreclinical[486]
Iadademstat (ORY-1001)Breast, AML, SCLCPhase II[487,488]
EUDRACT 2013-002447-29, NCT05420636, NCT05546580
Sonic HedgehogVismodegib (GDC-0449)Pancreatic, BCC, gastric, ovarianFDA-approved for BCC[496,497,498,499,500,501,502]
NCT00607724, NCT00833417, NCT03052478,
NCT00739661,
NCT01088815
Sonidegib (LDE225)CML, BCC, NSCLC, SCLC, medulloblastoma, breastFDA-approved for BCC[503,504,505,506,507,508]
NCT01327053, NCT01579929, NCT02027376,
NCT01125800,
NCT01456676
Glasdegib (PF-04449913)AMLFDA-approved for AML[509,510,511,512]
NCT03416179
Cyclopamine BreastPreclinical[513,514]
IPI-269609PancreaticPreclinical[515]
Saridegib (IPI-926)Medulloblastoma, chondrosarcoma, HNSCCPhase I[495,516,517,518,519,520,521]
NCT01255800,
NCT01383538,
NCT01310816
BMS-833923 (XL139)Esophageal, ovarian, AML, CMLPhase I[522,523,524,525]
NCT01218477
Taladegib (LY2940680, ENV-101)Medulloblastoma, BCCPhase II[526,527]
NCT01919398, NCT02784795,
NCT01226485,
NCT05199584
Hh003CRC, pancreaticPreclinical[528]
ItraconazoleMedulloblastoma, CRC, breast, biliary tractFDA-approved anti-fungal[529,530,531,532,533,534,535,536,537]
NCT00769600
ATOSCLC, Ewing sarcoma, pancreaticFDA-approved for acute promyelocytic leukemia[538,539,540,541,542]
GANT61Melanoma, pancreatic, HCCPreclinical[492,494,543,544,545]
MEDI-5304CRC, pancreaticPreclinical[546]
5E1CRC, pancreatic, breastPreclinical[546,547,548,549]
α-Ptch1PancreaticPreclinical[550]

Author Contributions

M.R.M.: conceptualization, data curation, writing—original draft, review and editing. O.L.W.: writing—original draft. R.P.A.: writing—original draft. W.F.: writing—olriginal draft. P.M.: conceptualization, supervision, writing—review and editing, project administration. All authors have read and agreed to the published version of the manuscript.

Funding

The article was funded by grant support to P.M. from the Canadian Institutes of Health Research (CHIR, PJT 162313). M.R.M is funded by a Nova Scotia Graduate Scholarship. O.L.W. is funded by a Killam Predoctoral Scholarship and the President’s Award. R.A.P. is a trainee in the Cancer Research Training Program of the Beatrice Hunter Cancer Research Institute, with funds generously provided by the Canadian Cancer Society’s JD Irving, Limited—Excellence in Cancer Research Fund.

Acknowledgments

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection; analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish.

References

  1. Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective Identification of Tumorigenic Breast Cancer Cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef] [PubMed]
  2. Ayob, A.Z.; Ramasamy, T.S. Cancer Stem Cells as Key Drivers of Tumor Progression. J. Biomed. Sci. 2018, 25, 20. [Google Scholar] [CrossRef]
  3. Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 Is a Marker of Normal and Malignant Human Mammary Stem Cells and a Predictor of Poor Clinical Outcome. Cell Stem Cell 2007, 1, 555–567. [Google Scholar] [CrossRef]
  4. Chen, K.; Huang, Y.H.; Chen, J.L. Understanding and Targeting Cancer Stem Cells: Therapeutic Implications and Challenges. Acta Pharmacol. Sin. 2013, 34, 732–740. [Google Scholar] [CrossRef] [PubMed]
  5. Ishizawa, K.; Rasheed, Z.A.; Karisch, R.; Wang, Q.; Kowalski, J.; Susky, E.; Pereira, K.; Karamboulas, C.; Moghal, N.; Rajeshkumar, N.V.; et al. Tumor-Initiating Cells Are Rare in Many Human Tumors. Cell Stem Cell 2010, 7, 279. [Google Scholar] [CrossRef]
  6. Toledo-Guzmán, M.E.; Bigoni-Ordóñez, G.D.; Hernández, M.I.; Ortiz-Sánchez, E. Cancer Stem Cell Impact on Clinical Oncology. World J. Stem Cells 2018, 10, 183. [Google Scholar] [CrossRef]
  7. Kurth, I.; Hein, L.; Mäbert, K.; Peitzsch, C.; Koi, L.; Cojoc, M.; Kunz-Schughart, L.; Baumann, M.; Dubrovska, A. Cancer Stem Cell Related Markers of Radioresistance in Head and Neck Squamous Cell Carcinoma. Oncotarget 2015, 6, 34494–34509. [Google Scholar] [CrossRef]
  8. Canino, C.; Luo, Y.Y.; Marcato, P.; Blandino, G.; Pass, H.I.; Cioce, M. A STAT3-NFkB/DDIT3/CEBPβ Axis Modulates ALDH1A3 Expression in Chemoresistant Cell Subpopulations. Oncotarget 2015, 6, 12637. [Google Scholar] [CrossRef] [PubMed]
  9. Nguyen, P.H.; Giraud, J.; Chambonnier, L.; Dubus, P.; Wittkop, L.; Belleannee, G.; Collet, D.; Soubeyran, I.; Evrard, S.; Rousseau, B.; et al. Characterization of Biomarkers of Tumorigenic and Chemoresistant Cancer Stem Cells in Human Gastric Carcinoma. Clin. Cancer Res. 2017, 23, 1586–1597. [Google Scholar] [CrossRef]
  10. Alvero, A.B.; Chen, R.; Fu, H.H.; Montagna, M.; Schwartz, P.E.; Rutherford, T.; Silasi, D.A.; Steffensen, K.D.; Waldstrom, M.; Visintin, I.; et al. Molecular Phenotyping of Human Ovarian Cancer Stem Cells Unravels the Mechanisms for Repair and Chemoresistance. Cell Cycle 2009, 8, 158–166. [Google Scholar] [CrossRef]
  11. Phi, L.T.H.; Sari, I.N.; Yang, Y.G.; Lee, S.H.; Jun, N.; Kim, K.S.; Lee, Y.K.; Kwon, H.Y. Cancer Stem Cells (CSCs) in Drug Resistance and Their Therapeutic Implications in Cancer Treatment. Stem Cells Int. 2018, 2018, 5416923. [Google Scholar] [CrossRef] [PubMed]
  12. Yamashita, D.; Minata, M.; Ibrahim, A.N.; Yamaguchi, S.; Coviello, V.; Bernstock, J.D.; Harada, S.; Cerione, R.A.; Tannous, B.A.; la Motta, C.; et al. Identification of ALDH1A3 as a Viable Therapeutic Target in Breast Cancer Metastasis-Initiating Cells. Mol. Cancer Ther. 2020, 19, 1134–1147. [Google Scholar] [CrossRef] [PubMed]
  13. Wei, P.L.; Prince, G.M.S.H.; Batzorig, U.; Huang, C.Y.; Chang, Y.J. ALDH2 Promotes Cancer Stemness and Metastasis in Colorectal Cancer through Activating β-Catenin Signaling. J. Cell Biochem. 2023, 124, 907–920. [Google Scholar] [CrossRef] [PubMed]
  14. Shiozawa, Y.; Nie, B.; Pienta, K.J.; Morgan, T.M.; Taichman, R.S. Cancer Stem Cells and Their Role in Metastasis. Pharmacol. Ther. 2013, 138, 285. [Google Scholar] [CrossRef] [PubMed]
  15. Horst, D.; Kriegl, L.; Engel, J.; Kirchner, T.; Jung, A. Prognostic Significance of the Cancer Stem Cell Markers CD133, CD44, and CD166 in Colorectal Cancer. Cancer Investig. 2009, 27, 844–850. [Google Scholar] [CrossRef] [PubMed]
  16. Nie, S.; Qian, X.; Shi, M.; Li, H.; Peng, C.; Ding, X.; Zhang, S.; Zhang, B.; Xu, G.; Lv, Y.; et al. ALDH1A3 Accelerates Pancreatic Cancer Metastasis by Promoting Glucose Metabolism. Front. Oncol. 2020, 10, 915. [Google Scholar] [CrossRef] [PubMed]
  17. Peitzsch, C.; Tyutyunnykova, A.; Pantel, K.; Dubrovska, A. Cancer Stem Cells: The Root of Tumor Recurrence and Metastases. Semin. Cancer Biol. 2017, 44, 10–24. [Google Scholar] [CrossRef]
  18. Steinbichler, T.B.; Savic, D.; Dudás, J.; Kvitsaridze, I.; Skvortsov, S.; Riechelmann, H.; Skvortsova, I.I. Cancer Stem Cells and Their Unique Role in Metastatic Spread. Semin. Cancer Biol. 2020, 60, 148–156. [Google Scholar] [CrossRef] [PubMed]
  19. Chen, W.; Dong, J.; Haiech, J.; Kilhoffer, M.C.; Zeniou, M. Cancer Stem Cell Quiescence and Plasticity as Major Challenges in Cancer Therapy. Stem Cells Int. 2016, 2016, 1740936. [Google Scholar] [CrossRef]
  20. Dembinski, J.L.; Krauss, S. Characterization and Functional Analysis of a Slow Cycling Stem Cell-like Subpopulation in Pancreas Adenocarcinoma. Clin. Exp. Metastasis 2009, 26, 611–623. [Google Scholar] [CrossRef]
  21. Chen, J.; Li, Y.; Yu, T.S.; McKay, R.M.; Burns, D.K.; Kernie, S.G.; Parada, L.F. A Restricted Cell Population Propagates Glioblastoma Growth after Chemotherapy. Nature 2012, 488, 522–526. [Google Scholar] [CrossRef] [PubMed]
  22. Marchitti, S.A.; Brocker, C.; Stagos, D.; Vasiliou, V. Non-P450 Aldehyde Oxidizing Enzymes: The Aldehyde Dehydrogenase Superfamily. Expert Opin. Drug Metab. Toxicol. 2008, 4, 697–720. [Google Scholar] [CrossRef] [PubMed]
  23. Marcato, P.; Dean, C.A.; Giacomantonio, C.A.; Lee, P.W.K. Aldehyde Dehydrogenase: Its Role as a Cancer Stem Cell Marker Comes down to the Specific Isoform. Cell Cycle 2011, 10, 1378–1384. [Google Scholar] [CrossRef] [PubMed]
  24. Vasillou, V.; Pappa, A.; Estey, T. Role of Human Aldehyde Dehydrogenases in Endobiotic and Xenobiotic Metabolism. Drug Metab. Rev. 2004, 36, 279–299. [Google Scholar] [CrossRef] [PubMed]
  25. Lalevée, S.; Anno, Y.N.; Chatagnon, A.; Samarut, E.; Poch, O.; Laudet, V.; Benoit, G.; Lecompte, O.; Rochette-Egly, C. Genome-Wide in Silico Identification of New Conserved and Functional Retinoic Acid Receptor Response Elements (Direct Repeats Separated by 5 Bp). J. Biol. Chem. 2011, 286, 33322–33334. [Google Scholar] [CrossRef] [PubMed]
  26. Coyle, K.M.; Sultan, M.; Thomas, M.L.; Vaghar-Kashani, A.; Marcato, P. Retinoid Signaling in Cancer and Its Promise for Therapy. J. Carcinog. Mutagen. 2013, 7, 16–18. [Google Scholar] [CrossRef]
  27. Zhou, L.; Sheng, D.; Wang, D.; Ma, W.; Deng, Q.; Deng, L.; Liu, S. Identification of Cancer-Type Specific Expression Patterns for Active Aldehyde Dehydrogenase (ALDH) Isoforms in ALDEFLUOR Assay. Cell Biol. Toxicol. 2019, 35, 161–177. [Google Scholar] [CrossRef] [PubMed]
  28. Kato, H.; Izumi, K.; Saito, T.; Ohnuki, H.; Terada, M.; Kawano, Y.; Nozawa-Inoue, K.; Saito, C.; Maeda, T. Distinct Expression Patterns and Roles of Aldehyde Dehydrogenases in Normal Oral Mucosa Keratinocytes: Differential Inhibitory Effects of a Pharmacological Inhibitor and RNAi-Mediated Knockdown on Cellular Phenotype and Epithelial Morphology. Histochem. Cell Biol. 2013, 139, 847–862. [Google Scholar] [CrossRef] [PubMed]
  29. Luo, Y.; Dallaglio, K.; Chen, Y.; Robinson, W.A.; Robinson, S.E.; McCarter, M.D.; Wang, J.; Gonzalez, R.; Thompson, D.C.; Norris, D.A.; et al. ALDH1A Isozymes Are Markers of Human Melanoma Stem Cells and Potential Therapeutic Targets. Stem Cells 2012, 30, 2100–2113. [Google Scholar] [CrossRef]
  30. Li, T.; Su, Y.; Mei, Y.; Leng, Q.; Leng, B.; Liu, Z.; Stass, S.A.; Jiang, F. ALDH1A1 Is a Marker for Malignant Prostate Stem Cells and Predictor of Prostate Cancer Patients’ Outcome. Lab. Investig. 2010, 90, 234–244. [Google Scholar] [CrossRef]
  31. Marcato, P.; Dean, C.A.; Da, P.; Araslanova, R.; Gillis, M.; Joshi, M.; Helyer, L.; Pan, L.; Leidal, A.; Gujar, S.; et al. Aldehyde Dehydrogenase Activity of Breast Cancer Stem Cells Is Primarily Due To Isoform ALDH1A3 and Its Expression Is Predictive of Metastasis. Stem Cells 2011, 29, 32–45. [Google Scholar] [CrossRef] [PubMed]
  32. Huang, E.H.; Hynes, M.J.; Zhang, T.; Ginestier, C.; Dontu, G.; Appelman, H.; Fields, J.Z.; Wicha, M.S.; Boman, B.M. Aldehyde Dehydrogenase 1 Is a Marker for Normal and Malignant Human Colonic Stem Cells (SC) and Tracks SC Overpopulation during Colon Tumorigenesis. Cancer Res. 2009, 69, 3382–3389. [Google Scholar] [CrossRef]
  33. Terzuoli, E.; Bellan, C.; Aversa, S.; Ciccone, V.; Morbidelli, L.; Giachetti, A.; Donnini, S.; Ziche, M. ALDH3A1 Overexpression in Melanoma and Lung Tumors Drives Cancer Stem Cell Expansion, Impairing Immune Surveillance through Enhanced PD-L1 Output. Cancers 2019, 11, 1963. [Google Scholar] [CrossRef]
  34. Chen, L.; Wu, M.; Ji, C.; Yuan, M.; Liu, C.; Yin, Q. Silencing Transcription Factor FOXM1 Represses Proliferation, Migration, and Invasion While Inducing Apoptosis of Liver Cancer Stem Cells by Regulating the Expression of ALDH2. IUBMB Life 2020, 72, 285–295. [Google Scholar] [CrossRef] [PubMed]
  35. Xu, S.L.; Liu, S.; Cui, W.; Shi, Y.; Liu, Q.; Duan, J.J.; Yu, S.C.; Zhang, X.; Cui, Y.H.; Kung, H.F.; et al. Aldehyde Dehydrogenase 1A1 Circumscribes High Invasive Glioma Cells and Predicts Poor Prognosis. Am. J. Cancer Res. 2015, 5, 1471. [Google Scholar] [PubMed]
  36. Mao, P.; Joshi, K.; Li, J.; Kim, S.H.; Li, P.; Santana-Santos, L.; Luthra, S.; Chandran, U.R.; Benos, P.V.; Smith, L.; et al. Mesenchymal Glioma Stem Cells Are Maintained by Activated Glycolytic Metabolism Involving Aldehyde Dehydrogenase 1A3. Proc. Natl. Acad. Sci. USA 2013, 110, 8644–8649. [Google Scholar] [CrossRef] [PubMed]
  37. Kawakami, R.; Mashima, T.; Kawata, N.; Kumagai, K.; Migita, T.; Sano, T.; Mizunuma, N.; Yamaguchi, K.; Seimiya, H. ALDH1A3-MTOR Axis as a Therapeutic Target for Anticancer Drug-Tolerant Persister Cells in Gastric Cancer. Cancer Sci. 2020, 111, 962–973. [Google Scholar] [CrossRef] [PubMed]
  38. Yang, L.; Ren, Y.; Yu, X.; Qian, F.; Bian, B.S.J.; Xiao, H.L.; Wang, W.G.; Xu, S.L.; Yang, J.; Cui, W.; et al. ALDH1A1 Defines Invasive Cancer Stem-like Cells and Predicts Poor Prognosis in Patients with Esophageal Squamous Cell Carcinoma. Mod. Pathol. 2014, 27, 775–783. [Google Scholar] [CrossRef] [PubMed]
  39. Li, X.; Xu, Q.; Fu, X.; Luo, W. ALDH1A1 Overexpression Is Associated with the Progression and Prognosis in Gastric Cancer. BMC Cancer 2014, 14, 705. [Google Scholar] [CrossRef]
  40. Meng, E.; Mitra, A.; Tripathi, K.; Finan, M.A.; Scalici, J.; McClellan, S.; Da Silva, L.M.; Reed, E.; Shevde, L.A.; Palle, K.; et al. ALDH1A1 Maintains Ovarian Cancer Stem Cell-like Properties by Altered Regulation of Cell Cycle Checkpoint and DNA Repair Network Signaling. PLoS ONE 2014, 9, e107142. [Google Scholar] [CrossRef]
  41. Khoury, T.; Ademuyiwa, F.O.; Chandraseekhar, R.; Jabbour, M.; Deleo, A.; Ferrone, S.; Wang, Y.; Wang, X. Aldehyde Dehydrogenase 1A1 Expression in Breast Cancer Is Associated with Stage, Triple Negativity, and Outcome to Neoadjuvant Chemotherapy. Mod. Pathol. 2012, 25, 388–397. [Google Scholar] [CrossRef]
  42. Kulsum, S.; Sudheendra, H.V.; Pandian, R.; Ravindra, D.R.; Siddappa, G.; Nisheena, R.; Chevour, P.; Ramachandran, B.; Sagar, M.; Jayaprakash, A.; et al. Cancer Stem Cell Mediated Acquired Chemoresistance in Head and Neck Cancer Can Be Abrogated by Aldehyde Dehydrogenase 1 A1 Inhibition. Mol. Carcinog. 2017, 56, 694–711. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, S.M.; Yasgar, A.; Miller, B.; Lal-Nag, M.; Brimacombe, K.; Hu, X.; Sun, H.; Wang, A.; Xu, X.; Nguyen, K.; et al. Discovery of NCT-501, a Potent and Selective Theophylline-Based Inhibitor of Aldehyde Dehydrogenase 1A1 (ALDH1A1). J. Med. Chem. 2015, 58, 5967. [Google Scholar] [CrossRef] [PubMed]
  44. Yasgar, A.; Titus, S.A.; Wang, Y.; Danchik, C.; Yang, S.M.; Vasiliou, V.; Jadhav, A.; Maloney, D.J.; Simeonov, A.; Martinez, N.J. A High-Content Assay Enables the Automated Screening and Identification of Small Molecules with Specific ALDH1A1-Inhibitory Activity. PLoS ONE 2017, 12, e0170937. [Google Scholar] [CrossRef] [PubMed]
  45. Nwani, N.G.; Condello, S.; Wang, Y.; Swetzig, W.M.; Barber, E.; Hurley, T.; Matei, D. A Novel ALDH1A1 Inhibitor Targets Cells with Stem Cell Characteristics in Ovarian Cancer. Cancers 2019, 11, 502. [Google Scholar] [CrossRef] [PubMed]
  46. Muralikrishnan, V.; Fang, F.; Given, T.C.; Podicheti, R.; Chtcherbinine, M.; Metcalfe, T.X.; Sriramkumar, S.; O’Hagan, H.M.; Hurley, T.D.; Nephew, K.P. A Novel ALDH1A1 Inhibitor Blocks Platinum-Induced Senescence and Stemness in Ovarian Cancer. Cancers 2022, 14, 3437. [Google Scholar] [CrossRef] [PubMed]
  47. Mclean, M.E.; Maclean, M.R.; Cahill, H.F.; Arun, R.P.; Walker, O.L.; Wasson, M.-C.D.; Fernando, W.; Ponnusamy, P.; Mclean, M.E.; Maclean, M.R.; et al. The Expanding Role of Cancer Stem Cell Marker ALDH1A3 in Cancer and Beyond. Cancers 2023, 15, 492. [Google Scholar] [CrossRef] [PubMed]
  48. Durinikova, E.; Kozovska, Z.; Poturnajova, M.; Plava, J.; Cierna, Z.; Babelova, A.; Bohovic, R.; Schmidtova, S.; Tomas, M.; Kucerova, L.; et al. ALDH1A3 Upregulation and Spontaneous Metastasis Formation Is Associated with Acquired Chemoresistance in Colorectal Cancer Cells. BMC Cancer 2018, 18, 848. [Google Scholar] [CrossRef] [PubMed]
  49. Marcato, P.; Dean, C.A.; Liu, R.Z.; Coyle, K.M.; Bydoun, M.; Wallace, M.; Clements, D.; Turner, C.; Mathenge, E.G.; Gujar, S.A.; et al. Aldehyde Dehydrogenase 1A3 Influences Breast Cancer Progression via Differential Retinoic Acid Signaling. Mol. Oncol. 2015, 9, 17–31. [Google Scholar] [CrossRef]
  50. Cheng, P.; Wang, J.; Waghmare, I.; Sartini, S.; Coviello, V.; Zhang, Z.; Kim, S.H.; Mohyeldin, A.; Pavlyukov, M.S.; Minata, M.; et al. FOXD1-ALDH1A3 Signaling Is a Determinant for the Self-Renewal and Tumorigenicity of Mesenchymal Glioma Stem Cells. Cancer Res. 2016, 76, 7219–7230. [Google Scholar] [CrossRef]
  51. Gelardi, E.L.M.; Colombo, G.; Picarazzi, F.; Ferraris, D.M.; Mangione, A.; Petrarolo, G.; Aronica, E.; Rizzi, M.; Mori, M.; La Motta, C.; et al. A Selective Competitive Inhibitor of Aldehyde Dehydrogenase 1A3 Hinders Cancer Cell Growth, Invasiveness and Stemness In Vitro. Cancers 2021, 13, 356. [Google Scholar] [CrossRef] [PubMed]
  52. Li, J.; Garavaglia, S.; Ye, Z.; Moretti, A.; Belyaeva, O.V.; Beiser, A.; Ibrahim, M.; Wilk, A.; McClellan, S.; Klyuyeva, A.V.; et al. A Specific Inhibitor of ALDH1A3 Regulates Retinoic Acid Biosynthesis in Glioma Stem Cells. Commun. Biol. 2021, 4, 1420. [Google Scholar] [CrossRef] [PubMed]
  53. Duan, J.-J.; Wang, D.; Cai, J.; Chen, J.; Zheng, X.-X.; Chen, T.; Wang, J.; Zhang, X.; Yang, Q.-K.; Yu, S.-C. An Aldehyde Dehydrogenase 1A3 Inhibitor Attenuates the Metastasis of Human Colorectal Cancer. Cancer Lett. 2022, 536, 215662. [Google Scholar] [CrossRef] [PubMed]
  54. Tran, T.O.; Vo, T.H.; Lam, L.H.T.; Le, N.Q.K. ALDH2 as a Potential Stem Cell-Related Biomarker in Lung Adenocarcinoma: Comprehensive Multi-Omics Analysis. Comput. Struct. Biotechnol. J. 2023, 21, 1921. [Google Scholar] [CrossRef] [PubMed]
  55. Arolfo, M.P.; Overstreet, D.H.; Yao, L.; Fan, P.; Lawrence, A.J.; Tao, G.; Keung, W.M.; Vallee, B.L.; Olive, M.F.; Gass, J.T.; et al. Suppression of Heavy Drinking and Alcohol Seeking by a Selective ALDH-2 Inhibitor. Alcohol. Clin. Exp. Res. 2009, 33, 1935. [Google Scholar] [CrossRef] [PubMed]
  56. Lowe, E.D.; Gao, G.Y.; Johnson, L.N.; Wing, M.K. Structure of Daidzin, a Naturally Occurring Anti-Alcohol-Addiction Agent, in Complex with Human Mitochondrial Aldehyde Dehydrogenase. J. Med. Chem. 2008, 51, 4482–4487. [Google Scholar] [CrossRef] [PubMed]
  57. Keung, W.M.; Vallee, B.L. Daidzin and Daidzein Suppress Free-Choice Ethanol Intake by Syrian Golden Hamsters. Proc. Natl. Acad. Sci. USA 1993, 90, 10008–10012. [Google Scholar] [CrossRef] [PubMed]
  58. Hu, G.; Chong, R.A.; Yang, Q.; Wei, Y.; Blanco, M.A.; Li, F.; Reiss, M.; Au, J.L.S.; Haffty, B.G.; Kang, Y. MTDH Activation by 8q22 Genomic Gain Promotes Chemoresistance and Metastasis of Poor-Prognosis Breast Cancer. Cancer Cell 2009, 15, 9–20. [Google Scholar] [CrossRef]
  59. Sládek, N.E.; Kollander, R.; Sreerama, L.; Kiang, D.T. Cellular Levels of Aldehyde Dehydrogenases (ALDH1A1 and ALDH3A1) as Predictors of Therapeutic Responses to Cyclophosphamide-Based Chemotherapy of Breast Cancer: A Retrospective Study. Cancer Chemother. Pharmacol. 2002, 49, 309–321. [Google Scholar] [CrossRef]
  60. Okazaki, S.; Shintani, S.; Hirata, Y.; Suina, K.; Semba, T.; Yamasaki, J.; Umene, K.; Ishikawa, M.; Saya, H.; Nagano, O. Synthetic Lethality of the ALDH3A1 Inhibitor Dyclonine and XCT Inhibitors in Glutathione Deficiency-Resistant Cancer Cells. Oncotarget 2018, 9, 33832. [Google Scholar] [CrossRef]
  61. Parajuli, B.; Fishel, M.L.; Hurley, T.D. Selective ALDH3A1 Inhibition by Benzimidazole Analogues Increase Mafosfamide Sensitivity in Cancer Cells. J. Med. Chem. 2014, 57, 449–461. [Google Scholar] [CrossRef] [PubMed]
  62. Parajuli, B.; Georgiadis, T.M.; Fishel, M.L.; Hurley, T.D. Development of Selective Inhibitors for Human Aldehyde Dehydrogenase 3A1 (ALDH3A1) for the Enhancement of Cyclophosphamide Cytotoxicity. ChemBioChem 2014, 15, 701–712. [Google Scholar] [CrossRef] [PubMed]
  63. Khanna, M.; Chen, C.H.; Kimble-Hill, A.; Parajuli, B.; Perez-Miller, S.; Baskaran, S.; Kim, J.; Dria, K.; Vasiliou, V.; Mochly-Rosen, D.; et al. Discovery of a Novel Class of Covalent Inhibitor for Aldehyde Dehydrogenases. J. Biol. Chem. 2011, 286, 43486–43494. [Google Scholar] [CrossRef] [PubMed]
  64. Counihan, J.L.; Wiggenhorn, A.L.; Anderson, K.E.; Nomura, D.K. Chemoproteomics-Enabled Covalent Ligand Screening Reveals ALDH3A1 as a Lung Cancer Therapy Target. ACS Chem. Biol. 2018, 13, 1970–1977. [Google Scholar] [CrossRef] [PubMed]
  65. Koppaka, V.; Thompson, D.C.; Chen, Y.; Ellermann, M.; Nicolaou, K.C.; Juvonen, R.O.; Petersen, D.; Deitrich, R.A.; Hurley, T.D.; Vasiliou, V. Aldehyde Dehydrogenase Inhibitors: A Comprehensive Review of the Pharmacology, Mechanism of Action, Substrate Specificity, and Clinical Application. Pharmacol. Rev. 2012, 64, 520–539. [Google Scholar] [CrossRef] [PubMed]
  66. Morgan, C.A.; Parajuli, B.; Buchman, C.D.; Dria, K.; Hurley, T.D. N,N-Diethylaminobenzaldehyde (DEAB) as a Substrate and Mechanism-Based Inhibitor for Human ALDH Isoenzymes. Chem. Biol. Interact. 2015, 234, 18–28. [Google Scholar] [CrossRef] [PubMed]
  67. Yue, L.; Huang, Z.M.; Fong, S.; Leong, S.; Jakowatz, J.G.; Charruyer-Reinwald, A.; Wei, M.; Ghadially, R. Targeting ALDH1 to Decrease Tumorigenicity, Growth and Metastasis of Human Melanoma. Melanoma Res. 2015, 25, 138–148. [Google Scholar] [CrossRef] [PubMed]
  68. N,N Diethylaminobenzaldehyde Targets Aldehyde Dehydrogenase to Eradicate Human Pancreatic Cancer Cells. Available online: https://www.spandidos-publications.com/10.3892/etm.2020.8691 (accessed on 3 December 2023).
  69. Dinavahi, S.S.; Gowda, R.; Gowda, K.; Bazewicz, C.G.; Chirasani, V.R.; Battu, M.B.; Berg, A.; Dokholyan, N.V.; Amin, S.; Robertson, G.P. Development of a Novel Multi-Isoform ALDH Inhibitor Effective as an Antimelanoma Agent. Mol. Cancer Ther. 2020, 19, 447–459. [Google Scholar] [CrossRef] [PubMed]
  70. Canuto, R.A.; Muzio, G.; Salvo, R.A.; Maggiora, M.; Trombetta, A.; Chantepie, J.; Fournet, G.; Reichert, U.; Quash, G. The Effect of a Novel Irreversible Inhibitor of Aldehyde Dehydrogenases 1 and 3 on Tumor Cell Growth and Death. Chem. Biol. Interact. 2001, 130–132, 209–218. [Google Scholar] [CrossRef]
  71. Venton, G.; Pérez-Alea, M.; Baier, C.; Fournet, G.; Quash, G.; Labiad, Y.; Martin, G.; Sanderson, F.; Poullin, P.; Suchon, P.; et al. Aldehyde Dehydrogenases Inhibition Eradicates Leukemia Stem Cells While Sparing Normal Progenitors. Blood Cancer J. 2016, 6, e469. [Google Scholar] [CrossRef]
  72. Chefetz, I.; Grimley, E.; Yang, K.; Hong, L.; Vinogradova, E.V.; Suciu, R.; Kovalenko, I.; Karnak, D.; Morgan, C.A.; Chtcherbinine, M.; et al. A Pan-ALDH1A Inhibitor Induces Necroptosis in Ovarian Cancer Stem-like Cells. Cell Rep. 2019, 26, 3061–3075.e6. [Google Scholar] [CrossRef] [PubMed]
  73. Thomas, M.L.; de Antueno, R.; Coyle, K.M.; Sultan, M.; Cruickshank, B.M.; Giacomantonio, M.A.; Giacomantonio, C.A.; Duncan, R.; Marcato, P. Citral Reduces Breast Tumor Growth by Inhibiting the Cancer Stem Cell Marker ALDH1A3. Mol. Oncol. 2016, 10, 1485–1496. [Google Scholar] [CrossRef] [PubMed]
  74. Pattanayak, R.; Sagar, R.; Pal, A. Tracing the Journey of Disulfiram: From an Unintended Discovery to a Treatment Option for Alcoholism. J. Ment. Health Hum. Behav. 2015, 20, 41. [Google Scholar] [CrossRef]
  75. Wang, N.N.; Wang, L.H.; Li, Y.; Fu, S.Y.; Xue, X.; Jia, L.N.; Yuan, X.Z.; Wang, Y.T.; Tang, X.; Yang, J.Y.; et al. Targeting ALDH2 with Disulfiram/Copper Reverses the Resistance of Cancer Cells to Microtubule Inhibitors. Exp. Cell Res. 2018, 362, 72–82. [Google Scholar] [CrossRef] [PubMed]
  76. Litvinov, S.V.; Balzar, M.; Winter, M.J.; Bakker, H.A.M.; Briaire-De Bruijn, I.H.; Prins, F.; Fleuren, G.J.; Warnaar, S.O. Epithelial Cell Adhesion Molecule (Ep-CAM) Modulates Cell–Cell Interactions Mediated by Classic Cadherins. J. Cell Biol. 1997, 139, 1337–1348. [Google Scholar] [CrossRef] [PubMed]
  77. Munz, M.; Baeuerle, P.A.; Gires, O. The Emerging Role of EpCAM in Cancer and Stem Cell Signaling. Cancer Res. 2009, 69, 5627–5629. [Google Scholar] [CrossRef] [PubMed]
  78. Ogura, E.; Senzaki, H.; Yoshizawa, K.; Hioki, K.; Tsubura, A. Immunohistochemical Localization of Epithelial Glycoprotein EGP-2 and Carcinoembryonic Antigen in Normal Colonic Mucosa and Colorectal Tumors. Anticancer Res. 1998, 18, 3669–3675. [Google Scholar] [PubMed]
  79. Herlyn, D.; Herlyn, M.; Steplewski, Z.; Koprowski, H. Monoclonal Antibodies in Cell-Mediated Cytotoxicity against Human Melanoma and Colorectal Carcinoma. Eur. J. Immunol. 1979, 9, 657–659. [Google Scholar] [CrossRef] [PubMed]
  80. Went, P.T.; Lugli, A.; Meier, S.; Bundi, M.; Mirlacher, M.; Sauter, G.; Dirnhofer, S. Frequent EpCam Protein Expression in Human Carcinomas. Hum. Pathol. 2004, 35, 122–128. [Google Scholar] [CrossRef]
  81. Lee, C.C.; Yu, C.J.; Panda, S.S.; Chen, K.C.; Liang, K.H.; Huang, W.C.; Wang, Y.S.; Ho, P.C.; Wu, H.C. Epithelial Cell Adhesion Molecule (EpCAM) Regulates HGFR Signaling to Promote Colon Cancer Progression and Metastasis. J. Transl. Med. 2023, 21, 530. [Google Scholar] [CrossRef]
  82. Hiraga, T.; Ito, S.; Nakamura, H. EpCAM Expression in Breast Cancer Cells Is Associated with Enhanced Bone Metastasis Formation. Int. J. Cancer 2016, 138, 1698–1708. [Google Scholar] [CrossRef] [PubMed]
  83. Terris, B.; Cavard, C.; Perret, C. EpCAM, a New Marker for Cancer Stem Cells in Hepatocellular Carcinoma. J. Hepatol. 2010, 52, 280–281. [Google Scholar] [CrossRef]
  84. Yamashita, T.; Ji, J.; Budhu, A.; Forgues, M.; Yang, W.; Wang, H.Y.; Jia, H.; Ye, Q.; Qin, L.X.; Wauthier, E.; et al. EpCAM-Positive Hepatocellular Carcinoma Cells Are Tumor-Initiating Cells with Stem/Progenitor Cell Features. Gastroenterology 2009, 136, 1012–1024. [Google Scholar] [CrossRef] [PubMed]
  85. Zheng, X.; Fan, X.; Fu, B.; Zheng, M.; Zhang, A.; Zhong, K.; Yan, J.; Sun, R.; Tian, Z.; Wei, H. EpCAM Inhibition Sensitizes Chemoresistant Leukemia to Immune Surveillance. Cancer Res. 2017, 77, 482–493. [Google Scholar] [CrossRef]
  86. Tayama, S.; Motohara, T.; Narantuya, D.; Li, C.; Fujimoto, K.; Sakaguchi, I.; Tashiro, H.; Saya, H.; Nagano, O.; Katabuchi, H. The Impact of EpCAM Expression on Response to Chemotherapy and Clinical Outcomes in Patients with Epithelial Ovarian Cancer. Oncotarget 2017, 8, 44312. [Google Scholar] [CrossRef] [PubMed]
  87. Wei, X.; Dombkowski, D.; Meirelles, K.; Pieretti-Vanmarcke, R.; Szotek, P.P.; Chang, H.L.; Preffer, F.I.; Mueller, P.R.; Teixeira, J.; MacLaughlin, D.T.; et al. Müllerian Inhibiting Substance Preferentially Inhibits Stem/Progenitors in Human Ovarian Cancer Cell Lines Compared with Chemotherapeutics. Proc. Natl. Acad. Sci. USA 2010, 107, 18874–18879. [Google Scholar] [CrossRef]
  88. Ensinger, C.; Kremser, R.; Prommegger, R.; Spizzo, G.; Schmid, K.W. EpCAM Overexpression in Thyroid Carcinomas: A Histopathological Study of 121 Cases. J. Immunother. 2006, 29, 569–573. [Google Scholar] [CrossRef] [PubMed]
  89. Seligson, D.B.; Pantuck, A.J.; Liu, X.; Huang, Y.; Horvath, S.; Bui, M.H.T.; Han, K.R.; Correa, A.J.L.; Eeva, M.; Tze, S.; et al. Epithelial Cell Adhesion Molecule (KSA) Expression: Pathobiology and Its Role as an Independent Predictor of Survival in Renal Cell Carcinoma. Clin. Cancer Res. 2004, 10, 2659–2669. [Google Scholar] [CrossRef]
  90. Liao, M.-Y.; Lai, J.-K.; Kuo, M.Y.-P.; Lu, R.-M.; Lin, C.-W.; Cheng, P.-C.; Liang, K.-H.; Wu, H.-C. An Anti-EpCAM Antibody EpAb2-6 for the Treatment of Colon Cancer. Oncotarget 2015, 6, 24947. [Google Scholar] [CrossRef]
  91. Chen, H.N.; Liang, K.H.; Lai, J.K.; Lan, C.H.; Liao, M.Y.; Hung, S.H.; Chuang, Y.T.; Chen, K.C.; Tsuei, W.W.F.; Wu, H.C. EpCAM Signaling Promotes Tumor Progression and Protein Stability of PD-L1 through the EGFR Pathway. Cancer Res. 2020, 80, 5035–5050. [Google Scholar] [CrossRef]
  92. Naundorf, S.; Preithner, S.; Mayer, P.; Lippold, S.; Wolf, A.; Hanakam, F.; Fichtner, I.; Kufer, P.; Raum, T.; Riethmüller, G.; et al. In Vitro and in Vivo Activity of MT201, a Fully Human Monoclonal Antibody for Pancarcinoma Treatment. Int. J. Cancer 2002, 100, 101–110. [Google Scholar] [CrossRef] [PubMed]
  93. Schmidt, M.; Scheulen, M.E.; Dittrich, C.; Obrist, P.; Marschner, N.; Dirix, L.; Schmidt, M.; Rüttinger, D.; Schuler, M.; Reinhardt, C.; et al. An Open-Label, Randomized Phase II Study of Adecatumumab, a Fully Human Anti-EpCAM Antibody, as Monotherapy in Patients with Metastatic Breast Cancer. Ann. Oncol. 2010, 21, 275–282. [Google Scholar] [CrossRef] [PubMed]
  94. Marschner, N.; Rüttinger, D.; Zugmaier, G.; Nemere, G.; Lehmann, J.; Obrist, P.; Baeuerle, P.A.; Wolf, A.; Schmidt, M.; Abrahamsson, P.A.; et al. Phase II Study of the Human Anti-Epithelial Cell Adhesion Molecule Antibody Adecatumumab in Prostate Cancer Patients with Increasing Serum Levels of Prostate-Specific Antigen after Radical Prostatectomy. Urol. Int. 2010, 85, 386–395. [Google Scholar] [CrossRef]
  95. Sebastian, M.; Hanusch, C.; Schmidt, M.; Marschner, N.; Oruzio, D.; Wolf, C.; Reinhardt, C.; Eiermann, W.; Rüttinger, D.; Schuler, M. Safety and Antitumor Activity of 3-Weekly Anti-EpCAM Antibody Adecatumumab (MT201) in Combination with Docetaxel for Patients with Metastatic Breast Cancer: Results of a Multicenter Phase Ib Trial. J. Clin. Oncol. 2009, 27 (Suppl. S15), 1009. [Google Scholar] [CrossRef]
  96. Study Details|Safety of AM-928 Infusion in Advanced Solid Tumors|ClinicalTrials.gov. Available online: https://clinicaltrials.gov/study/NCT05687682 (accessed on 5 January 2024).
  97. Tian, Z.; Liu, M.; Zhang, Y.; Wang, X. Bispecific T Cell Engagers: An Emerging Therapy for Management of Hematologic Malignancies. J. Hematol. Oncol. 2021, 14, 75. [Google Scholar] [CrossRef] [PubMed]
  98. Cioffi, M.; Dorado, J.; Baeuerle, P.A.; Heeschen, C. EpCAM/CD3-Bispecific T-Cell Engaging Antibody MT110 Eliminates Primary Human Pancreatic Cancer Stem Cells. Clin. Cancer Res. 2012, 18, 465–474. [Google Scholar] [CrossRef]
  99. English, D.P.; Bellone, S.; Schwab, C.L.; Roque, D.M.; Lopez, S.; Bortolomai, I.; Cocco, E.; Bonazzoli, E.; Chatterjee, S.; Ratner, E.; et al. Solitomab, an Epithelial Cell Adhesion Molecule/CD3 Bispecific Antibody (BiTE), Is Highly Active against Primary Chemotherapy-Resistant Ovarian Cancer Cell Lines in Vitro and Fresh Tumor Cells Ex Vivo. Cancer 2015, 121, 403–412. [Google Scholar] [CrossRef] [PubMed]
  100. Kebenko, M.; Goebeler, M.E.; Wolf, M.; Hasenburg, A.; Seggewiss-Bernhardt, R.; Ritter, B.; Rautenberg, B.; Atanackovic, D.; Kratzer, A.; Rottman, J.B.; et al. A Multicenter Phase 1 Study of Solitomab (MT110, AMG 110), a Bispecific EpCAM/CD3 T-Cell Engager (BiTE®) Antibody Construct, in Patients with Refractory Solid Tumors. Oncoimmunology 2018, 7, 1450710. [Google Scholar] [CrossRef]
  101. Linke, R.; Klein, A.; Seimetz, D. Catumaxomab: Clinical Development and Future Directions. MAbs 2010, 2, 129. [Google Scholar] [CrossRef]
  102. Golubovskaya, V.; Sienkiewicz, J.; Sun, J.; Huang, Y.; Hu, L.; Zhou, H.; Harto, H.; Xu, S.; Berahovich, R.; Bodmer, W.; et al. MRNA-Lipid Nanoparticle (LNP) Delivery of Humanized EpCAM-CD3 Bispecific Antibody Significantly Blocks Colorectal Cancer Tumor Growth. Cancers 2023, 15, 2860. [Google Scholar] [CrossRef]
  103. Ang, W.X.; Li, Z.; Chi, Z.; Du, S.-H.; Chen, C.; Tay, J.C.K.; Toh, H.C.; Connolly, J.E.; Xu, X.H.; Wang, S.; et al. Intraperitoneal Immunotherapy with T Cells Stably and Transiently Expressing Anti-EpCAM CAR in Xenograft Models of Peritoneal Carcinomatosis. Oncotarget 2017, 8, 13545–13559. [Google Scholar] [CrossRef] [PubMed]
  104. Deng, Z.; Wu, Y.; Ma, W.; Zhang, S.; Zhang, Y.Q. Adoptive T-Cell Therapy of Prostate Cancer Targeting the Cancer Stem Cell Antigen EpCAM. BMC Immunol. 2015, 16, 1. [Google Scholar] [CrossRef] [PubMed]
  105. Xu, T.; Karschnia, P.; Cadilha, B.L.; Dede, S.; Lorenz, M.; Seewaldt, N.; Nikolaishvili, E.; Müller, K.; Blobner, J.; Teske, N.; et al. In Vivo Dynamics and Anti-Tumor Effects of EpCAM-Directed CAR T-Cells against Brain Metastases from Lung Cancer. Oncoimmunology 2023, 12, 2163781. [Google Scholar] [CrossRef] [PubMed]
  106. Yang, Y.; McCloskey, J.E.; Yang, H.; Puc, J.; Alcaina, Y.; Vedvyas, Y.; Gomez Gallegos, A.A.; Ortiz-Sánchez, E.; de Stanchina, E.; Min, I.M.; et al. Bispecific CAR T Cells against EpCAM and Inducible ICAM-1 Overcome Antigen Heterogeneity and Generate Superior Anti-Tumor Responses. Cancer Immunol. Res. 2021, 9, 1158. [Google Scholar] [CrossRef] [PubMed]
  107. Gardner, R.; Wu, D.; Cherian, S.; Fang, M.; Hanafi, L.A.; Finney, O.; Smithers, H.; Jensen, M.C.; Riddell, S.R.; Maloney, D.G.; et al. Acquisition of a CD19-Negative Myeloid Phenotype Allows Immune Escape of MLL-Rearranged B-ALL from CD19 CAR-T-Cell Therapy. Blood 2016, 127, 2406–2410. [Google Scholar] [CrossRef] [PubMed]
  108. Luo, T.; Fang, W.; Lu, Z.; Tong, C.; Zhang, H.; Ai, G.; Wang, S. EpCAM CAR T (IMC001) for the Treatment of Advanced GI Cancers. J. Clin. Oncol. 2023, 41 (Suppl. S16), 4034. [Google Scholar] [CrossRef]
  109. Entwistle, J.; Kowalski, M.; Brown, J.; Cizeau, J.; MacDonald, G.C. The Preclinical and Clinical Evaluation of VB6- 845: An Immunotoxin with a de-Immunized Payload for the Systemic Treatment of Solid Tumors. In Antibody-Drug Conjugates and Immunotoxins: From Pre-Clinical Development to Therapeutic Applications; Springer: New York, NY, USA, 2013; pp. 349–367. [Google Scholar] [CrossRef]
  110. Cizeau, J.; Grenkow, D.M.; Brown, J.G.; Entwistle, J.; MacDonald, G.C. Engineering and Biological Characterization of VB6-845, an Anti-EpCAM Immunotoxin Containing a t-Cell Epitope-Depleted Variant of the Plant Toxin Bouganin. J. Immunother. 2009, 32, 574–584. [Google Scholar] [CrossRef] [PubMed]
  111. Kowalski, M.; Brazas, L.; Zaretsky, R.; Rasamoelisolo, M.; MacDonald, G.; Cuthbert, W.; Glover, N. A Phase I Study of VB6–845, an Anti-EpCAM Fusion Protein Targeting Advanced Solid Tumors of Epithelial Origin: Preliminary Results. J. Clin. Oncol. 2008, 26 (Suppl. S15), 14663. [Google Scholar] [CrossRef]
  112. Brown, J.; Rasamoelisolo, M.; Spearman, M.; Bosc, D.; Cizeau, J.; Entwistle, J.; MacDonald, G.C. Preclinical Assessment of an Anti-EpCAM Immunotoxin: Locoregional Delivery Provides a Safer Alternative to Systemic Administration. Cancer Biother. Radiopharm. 2009, 24, 477–487. [Google Scholar] [CrossRef]
  113. Dillon, R.L.; Chooniedass, S.; Premsukh, A.; MacDonald, G.; Cizeau, J.; Adams, G.A. Abstract 614: VB4-845 Tumor Cell Killing in a Combination Study with the Anti-PD-1, Nivolumab. Cancer Res. 2017, 77 (Suppl. S13), 614. [Google Scholar] [CrossRef]
  114. Fitsialos, D.; Quenneville, J.; Rasamoelisolo, M.; Cross, M.; Glover, N.; MacDonald, G. A Phase I Study of VB4–845 in Patients with Advanced, Recurrent Head and Neck Cancer on a Weekly Dosing Scheme. J. Clin. Oncol. 2005, 23 (Suppl. S16), 5569. [Google Scholar] [CrossRef]
  115. MacDonald, G.C.; Rasamoelisolo, M.; Entwistle, J.; Cizeau, J.; Bosc, D.; Cuthbert, W.; Kowalski, M.; Spearman, M.; Glover, N. A Phase I Clinical Study of VB4-845: Weekly Intratumoral Administration of an Anti-EpCAM Recombinant Fusion Protein in Patients with Squamous Cell Carcinoma of the Head and Neck. Drug Des. Devel Ther. 2008, 2, 105. [Google Scholar] [CrossRef]
  116. Goldberg, I.P.; Lichtbroun, B.; Singer, E.A.; Ghodoussipour, S. Pharmacologic Therapies for Non-Muscle Invasive Bladder Cancer: Current and Future Treatments. Arch. Pharmacol. Ther. 2022, 4, 13. [Google Scholar] [PubMed]
  117. Lv, M.; Qiu, F.; Li, T.; Sun, Y.; Zhang, C.; Zhu, P.; Qi, X.; Wan, J.; Yang, K.; Zhang, K. Construction, Expression, and Characterization of a Recombinant Immunotoxin Targeting EpCAM. Mediat. Inflamm. 2015, 2015, 460264. [Google Scholar] [CrossRef] [PubMed]
  118. Tretter, J.Y.; Schorpp, K.; Luxenburger, E.; Trambauer, J.; Steiner, H.; Hadian, K.; Gires, O.; Niessing, D. A High-Content Screen for Small-Molecule Regulators of Epithelial Cell-Adhesion Molecule (EpCAM) Cleavage Yields a Robust Inhibitor. J. Biol. Chem. 2018, 293, 8994–9005. [Google Scholar] [CrossRef] [PubMed]
  119. Leblanc, R.; Kashyap, R.; Barral, K.; Egea-Jimenez, A.L.; Kovalskyy, D.; Feracci, M.; Garcia, M.; Derviaux, C.; Betzi, S.; Ghossoub, R.; et al. Pharmacological Inhibition of Syntenin PDZ2 Domain Impairs Breast Cancer Cell Activities and Exosome Loading with Syndecan and EpCAM Cargo. J. Extracell. Vesicles 2020, 10, e12039. [Google Scholar] [CrossRef] [PubMed]
  120. Moldenhauer, G.; Salnikov, A.V.; Lüttgau, S.; Herr, I.; Anderl, J.; Faulstich, H. Therapeutic Potential of Amanitin-Conjugated Anti-Epithelial Cell Adhesion Molecule Monoclonal Antibody Against Pancreatic Carcinoma. JNCI J. Natl. Cancer Inst. 2012, 104, 622–634. [Google Scholar] [CrossRef] [PubMed]
  121. Lakhin, A.V.; Tarantul, V.Z.; Gening, L.V. Aptamers: Problems, Solutions and Prospects. Acta Naturae 2013, 5, 34. [Google Scholar] [CrossRef] [PubMed]
  122. Xiang, D.; Zheng, C.; Zhou, S.F.; Qiao, S.; Tran, P.H.L.; Pu, C.; Li, Y.; Kong, L.; Kouzani, A.Z.; Lin, J.; et al. Superior Performance of Aptamer in Tumor Penetration over Antibody: Implication of Aptamer-Based Theranostics in Solid Tumors. Theranostics 2015, 5, 1083. [Google Scholar] [CrossRef]
  123. Subramanian, N.; Kanwar, J.R.; Athalya, P.K.; Janakiraman, N.; Khetan, V.; Kanwar, R.K.; Eluchuri, S.; Krishnakumar, S. EpCAM Aptamer Mediated Cancer Cell Specific Delivery of EpCAM SiRNA Using Polymeric Nanocomplex. J. Biomed. Sci. 2015, 22, 4. [Google Scholar] [CrossRef]
  124. Mitra, M.; Kandalam, M.; Rangasamy, J.; Shankar, B.; Maheswari, U.K.; Swaminathan, S.; Krishnakumar, S. Novel Epithelial Cell Adhesion Molecule Antibody Conjugated Polyethyleneimine-Capped Gold Nanoparticles for Enhanced and Targeted Small Interfering RNA Delivery to Retinoblastoma Cells. Mol. Vis. 2013, 19, 1029. [Google Scholar] [PubMed]
  125. Ponta, H.; Sherman, L.; Herrlich, P.A. CD44: From Adhesion Molecules to Signaling Regulators. Nat. Rev. Mol. Cell Biol. 2003, 4, 33–45. [Google Scholar] [CrossRef]
  126. Yu, Q.; Stamenkovic, I. Localization of Matrix Metalloproteinase 9 to the Cell Surface Provides a Mechanism for CD44-Mediated Tumor Invasion. Genes Dev. 1999, 13, 35. [Google Scholar] [CrossRef]
  127. Zöller, M. CD44: Can a Cancer-Initiating Cell Profit from an Abundantly Expressed Molecule? Nat. Rev. Cancer 2011, 11, 254–267. [Google Scholar] [CrossRef]
  128. Walcher, L.; Kistenmacher, A.K.; Suo, H.; Kitte, R.; Dluczek, S.; Strauß, A.; Blaudszun, A.R.; Yevsa, T.; Fricke, S.; Kossatz-Boehlert, U. Cancer Stem Cells—Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front. Immunol. 2020, 11, 539291. [Google Scholar] [CrossRef] [PubMed]
  129. Jin, L.; Hope, K.J.; Zhai, Q.; Smadja-Joffe, F.; Dick, J.E. Targeting of CD44 Eradicates Human Acute Myeloid Leukemic Stem Cells. Nat. Med. 2006, 12, 1167–1174. [Google Scholar] [CrossRef] [PubMed]
  130. Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The Epithelial-Mesenchymal Transition Generates Cells with Properties of Stem Cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [PubMed]
  131. Zhao, S.; Chen, C.; Chang, K.; Karnad, A.; Jagirdar, J.; Kumar, A.P.; Freeman, J.W. CD44 Expression Level and Isoform Contributes to Pancreatic Cancer Cell Plasticity, Invasiveness, and Response to Therapy. Clin. Cancer Res. 2016, 22, 5592–5604. [Google Scholar] [CrossRef] [PubMed]
  132. Takaishi, S.; Okumura, T.; Tu, S.; Wang, S.S.W.; Shibata, W.; Vigneshwaran, R.; Gordon, S.A.K.; Shimada, Y.; Wang, T.C. Identification of Gastric Cancer Stem Cells Using the Cell Surface Marker CD44. Stem Cells 2009, 27, 1006–1020. [Google Scholar] [CrossRef]
  133. Riechelmann, H.; Sauter, A.; Golze, W.; Hanft, G.; Schroen, C.; Hoermann, K.; Erhardt, T.; Gronau, S. Phase I Trial with the CD44v6-Targeting Immunoconjugate Bivatuzumab Mertansine in Head and Neck Squamous Cell Carcinoma. Oral. Oncol. 2008, 44, 823–829. [Google Scholar] [CrossRef]
  134. Li, L.; Hao, X.; Qin, J.; Tang, W.; He, F.; Smith, A.; Zhang, M.; Simeone, D.M.; Qiao, X.T.; Chen, Z.N.; et al. Antibody Against CD44s Inhibits Pancreatic Tumor Initiation and Postradiation Recurrence in Mice. Gastroenterology 2014, 146, 1108–1118.e12. [Google Scholar] [CrossRef]
  135. Uchino, M.; Kojima, H.; Wada, K.; Imada, M.; Onoda, F.; Satofuka, H.; Utsugi, T.; Murakami, Y. Nuclear β-Catenin and CD44 Upregulation Characterize Invasive Cell Populations in Non-Aggressive MCF-7 Breast Cancer Cells. BMC Cancer 2010, 10, 414. [Google Scholar] [CrossRef]
  136. Hellqvist, E.; Holm, F.; Mason, C.N.; Runza, V.; Weigand, S.; Sadarangani, A.; Jamieson, C.H.M. CD44 Monoclonal Antibody-Enhanced Clearance of Chronic Myeloid Leukemia Stem Cells from The Malignant Niche. Blood 2013, 122, 858. [Google Scholar] [CrossRef]
  137. Zhang, S.; Wu, C.C.N.; Fecteau, J.F.; Cui, B.; Chen, L.; Zhang, L.; Wu, R.; Rassenti, L.; Lao, F.; Weigand, S.; et al. Targeting Chronic Lymphocytic Leukemia Cells with a Humanized Monoclonal Antibody Specific for CD44. Proc. Natl. Acad. Sci. USA 2013, 110, 6127–6132. [Google Scholar] [CrossRef]
  138. Perez, A.; Neskey, D.M.; Wen, J.; Goodwin, J.W.; Slingerland, J.; Pereira, L.; Weigand, S.; Franzmann, E.J. Abstract 2521: Targeting CD44 in Head and Neck Squamous Cell Carcinoma (HNSCC) with a New Humanized Antibody RO5429083. Cancer Res. 2012, 72 (Suppl. S8), 2521. [Google Scholar] [CrossRef]
  139. Menke-van der Houven van Oordt, C.W.; Gomez-Roca, C.; van Herpen, C.; Coveler, A.L.; Mahalingam, D.; Verheul, H.M.W.; van der Graaf, W.T.A.; Christen, R.; Rüttinger, D.; Weigand, S.; et al. First-in-Human Phase I Clinical Trial of RG7356, an Anti-CD44 Humanized Antibody, in Patients with Advanced, CD44-Expressing Solid Tumors. Oncotarget 2016, 7, 80046. [Google Scholar] [CrossRef] [PubMed]
  140. Vey, N.; Delaunay, J.; Martinelli, G.; Fiedler, W.; Raffoux, E.; Prebet, T.; Gomez-Roca, C.; Papayannidis, C.; Kebenko, M.; Paschka, P.; et al. Phase I Clinical Study of RG7356, an Anti-CD44 Humanized Antibody, in Patients with Acute Myeloid Leukemia. Oncotarget 2016, 7, 32532–32542. [Google Scholar] [CrossRef]
  141. Mitsunaga, M.; Ogawa, M.; Kosaka, N.; Rosenblum, L.T.; Choyke, P.L.; Kobayashi, H. Cancer-Cell Selective in Vivo Near Infrared Photoimmunotherapy Targeting Specific Membrane Molecules. Nat. Med. 2011, 17, 1685–1691. [Google Scholar] [CrossRef] [PubMed]
  142. Nagaya, T.; Nakamura, Y.; Okuyama, S.; Ogata, F.; Maruoka, Y.; Choyke, P.L.; Allen, C.; Kobayashi, H. Syngeneic Mouse Models of Oral Cancer Are Effectively Targetedbyanti-Cd44-BasedNIR-PIT. Mol. Cancer Res. 2017, 15, 1667–1677. [Google Scholar] [CrossRef]
  143. Nagaya, T.; Friedman, J.; Maruoka, Y.; Ogata, F.; Okuyama, S.; Clavijo, P.E.; Choyke, P.L.; Allen, C.; Kobayashi, H. Host Immunity Following Near-Infrared Photoimmunotherapy Is Enhanced with PD-1 Checkpoint Blockade to Eradicate Established Antigenic Tumors. Cancer Immunol. Res. 2019, 7, 401–413. [Google Scholar] [CrossRef]
  144. Maruoka, Y.; Furusawa, A.; Okada, R.; Inagaki, F.; Fujimura, D.; Wakiyama, H.; Kato, T.; Nagaya, T.; Choyke, P.L.; Kobayashi, H. Combined CD44- And CD25-Targeted near-Infrared Photoimmunotherapy Selectively Kills Cancer and Regulatory T Cells in Syngeneic Mouse Cancer Models. Cancer Immunol. Res. 2020, 8, 345–355. [Google Scholar] [CrossRef] [PubMed]
  145. Waller, K.A.; Chin, K.E.; Jay, G.D.; Zhang, L.X.; Teeple, E.; McAllister, S.; Badger, G.J.; Schmidt, T.A.; Fleming, B.C. Intra-Articular Recombinant Human Proteoglycan 4 Mitigates Cartilage Damage after Destabilization of the Medial Meniscus in the Yucatan Minipig. Am. J. Sports Med. 2017, 45, 1512–1521. [Google Scholar] [CrossRef] [PubMed]
  146. Sarkar, A.; Chanda, A.; Regmi, S.C.; Karve, K.; Deng, L.; Jay, G.D.; Jirik, F.R.; Schmidt, T.A.; Bonni, S. Recombinant Human PRG4 (RhPRG4) Suppresses Breast Cancer Cell Invasion by Inhibiting TGFβ-Hyaluronan-CD44 Signaling Pathway. PLoS ONE 2019, 14, e0219697. [Google Scholar] [CrossRef] [PubMed]
  147. Transforming Growth Factor-β1 Induces EMT by the Transactivation of Epidermal Growth Factor Signaling through HA/CD44 in Lung and Breast Cancer Cells. Available online: https://www.spandidos-publications.com/10.3892/ijmm.2015.2222 (accessed on 18 December 2023).
  148. Rosenberg, J.E.; Bambury, R.M.; Van Allen, E.M.; Drabkin, H.A.; Lara, P.N.; Harzstark, A.L.; Wagle, N.; Figlin, R.A.; Smith, G.W.; Garraway, L.A.; et al. A Phase II Trial of AS1411 (a Novel Nucleolin-Targeted DNA Aptamer) in Metastatic Renal Cell Carcinoma. Investig. New Drugs 2014, 32, 178–187. [Google Scholar] [CrossRef] [PubMed]
  149. Iida, J.; Clancy, R.; Dorchak, J.; Somiari, R.I.; Somiari, S.; Cutler, M.L.; Mural, R.J.; Shriver, C.D. DNA Aptamers against Exon V10 of CD44 Inhibit Breast Cancer Cell Migration. PLoS ONE 2014, 9, e88712. [Google Scholar] [CrossRef]
  150. Zheng, J.; Zhao, S.; Yu, X.; Huang, S.; Liu, H.Y. Simultaneous Targeting of CD44 and EpCAM with a Bispecific Aptamer Effectively Inhibits Intraperitoneal Ovarian Cancer Growth. Theranostics 2017, 7, 1373. [Google Scholar] [CrossRef] [PubMed]
  151. Di Fusco, D.; Dinallo, V.; Marafini, I.; Figliuzzi, M.M.; Romano, B.; Monteleone, G. Antisense Oligonucleotide: Basic Concepts and Therapeutic Application in Inflammatory Bowel Disease. Front. Pharmacol. 2019, 10, 440751. [Google Scholar] [CrossRef] [PubMed]
  152. Xie, Z.; Choong, P.F.; Poon, L.F.; Zhou, J.; Khng, J.; Jasinghe, V.J.; Palaniyandi, S.; Chen, C.S. Inhibition of CD44 Expression in Hepatocellular Carcinoma Cells Enhances Apoptosis, Chemosensitivity, and Reduces Tumorigenesis and Invasion. Cancer Chemother. Pharmacol. 2008, 62, 949–957. [Google Scholar] [CrossRef] [PubMed]
  153. Bartolucci, D.; Pession, A.; Hrelia, P.; Tonelli, R. Precision Anti-Cancer Medicines by Oligonucleotide Therapeutics in Clinical Research Targeting Undruggable Proteins and Non-Coding RNAs. Pharmaceutics 2022, 14, 1453. [Google Scholar] [CrossRef]
  154. Liu, L.K.; Finzel, B.C. Fragment-Based Identification of an Inducible Binding Site on Cell Surface Receptor CD44 for the Design of Protein-Carbohydrate Interaction Inhibitors. J. Med. Chem. 2014, 57, 2714–2725. [Google Scholar] [CrossRef]
  155. Roy, S.; Kar, M.; Roy, S.; Padhi, S.; Kumar, A.; Thakur, S.; Akhter, Y.; Gatto, G.; Banerjee, B. Inhibition of CD44 Sensitizes Cisplatin-Resistance and Affects Wnt/β-Catenin Signaling in HNSCC Cells. Int. J. Biol. Macromol. 2020, 149, 501–512. [Google Scholar] [CrossRef] [PubMed]
  156. Espejo-Román, J.M.; Rubio-Ruiz, B.; Cano-Cortés, V.; Cruz-López, O.; Gonzalez-Resines, S.; Domene, C.; Conejo-García, A.; Sánchez-Martín, R.M. Selective Anticancer Therapy Based on a HA-CD44 Interaction Inhibitor Loaded on Polymeric Nanoparticles. Pharmaceutics 2022, 14, 788. [Google Scholar] [CrossRef] [PubMed]
  157. Wang, C.; Wang, Z.; Chen, C.; Fu, X.; Wang, J.; Fei, X.; Yan, X.; Xu, R. A Low MW Inhibitor of CD44 Dimerization for the Treatment of Glioblastoma. Br. J. Pharmacol. 2020, 177, 3009. [Google Scholar] [CrossRef]
  158. Finlayson, M. Modulation of CD44 Activity by A6-Peptide. Front. Immunol. 2015, 6, 132943. [Google Scholar] [CrossRef] [PubMed]
  159. Piotrowicz, R.S.; Damaj, B.B.; Hachicha, M.; Incardona, F.; Howell, S.B.; Finlayson, M. A6 Peptide Activates CD44 Adhesive Activity, Induces FAK and MEK Phosphorylation, and Inhibits the Migration and Metastasis of CD44-Expressing Cells. Mol. Cancer Ther. 2011, 10, 2072–2082. [Google Scholar] [CrossRef] [PubMed]
  160. Boyd, D.D.; Kim, S.J.; Wang, H.; Jones, T.R.; Gallick, G.E. A Urokinase-Derived Peptide (Å6) Increases Survival of Mice Bearing Orthotopically Grown Prostate Cancer and Reduces Lymph Node Metastasis. Am. J. Pathol. 2003, 162, 619–626. [Google Scholar] [CrossRef]
  161. Gu, W.; An, J.; Meng, H.; Yu, N.; Zhong, Y.; Meng, F.; Xu, Y.; Cornelissen, J.J.L.M.; Zhong, Z. CD44-Specific A6 Short Peptide Boosts Targetability and Anticancer Efficacy of Polymersomal Epirubicin to Orthotopic Human Multiple Myeloma. Adv. Mater. 2019, 31, 1904742. [Google Scholar] [CrossRef] [PubMed]
  162. Damaj, B.B.; Incardonna, F.; Piotrowicz, R.; Howell, S.B.; Finlayson, M. Abstract 5120: A6 Peptide Binds to CD44 and Inhibits Migration and Metastasis of CD44+ Cell Lines in in Vitro and in Vivo Studies. Cancer Res. 2010, 70 (Suppl. S8), 5120. [Google Scholar] [CrossRef]
  163. Gold, M.A.; Brady, W.E.; Lankes, H.A.; Rose, P.G.; Kelley, J.L.; De Geest, K.; Crispens, M.A.; Resnick, K.E.; Howell, S.B. A Phase II Study of a Urokinase-Derived Peptide (A6) in the Treatment of Persistent or Recurrent Epithelial Ovarian, Fallopian Tube, or Primary Peritoneal Carcinoma: A Gynecologic Oncology Group Study. Gynecol. Oncol. 2012, 125, 635–639. [Google Scholar] [CrossRef]
  164. Ghamande, S.A.; Silverman, M.H.; Huh, W.; Behbakht, K.; Ball, G.; Cuasay, L.; Würtz, S.O.; Brunner, N.; Gold, M.A. A Phase 2, Randomized, Double-Blind, Placebo-Controlled Trial of Clinical Activity and Safety of Subcutaneous Å6 in Women with Asymptomatic CA125 Progression after First-Line Chemotherapy of Epithelial Ovarian Cancer. Gynecol. Oncol. 2008, 111, 89–94. [Google Scholar] [CrossRef]
  165. Yazdian-Robati, R.; Amiri, E.; Kamali, H.; Khosravi, A.; Taghdisi, S.M.; Jaafari, M.R.; Mashreghi, M.; Moosavian, S.A. CD44-Specific Short Peptide A6 Boosts Cellular Uptake and Anticancer Efficacy of PEGylated Liposomal Doxorubicin in Vitro and in Vivo. Cancer Nanotechnol. 2023, 14, 84. [Google Scholar] [CrossRef]
  166. Bharti, R.; Dey, G.; Lin, F.; Lathia, J.; Reizes, O. CD55 in Cancer: Complementing Functions in a Non-Canonical Manner. Cancer Lett. 2022, 551, 215935. [Google Scholar] [CrossRef]
  167. Geller, A.; Yan, J. The Role of Membrane Bound Complement Regulatory Proteins in Tumor Development and Cancer Immunotherapy. Front. Immunol. 2019, 10, 440775. [Google Scholar] [CrossRef] [PubMed]
  168. Weng, Z.; Lin, J.; He, J.; Gao, L.; Lin, S.; Tsang, L.L.; Zhang, H.; He, X.; Wang, G.; Yang, X.; et al. Human Embryonic Stem Cell-Derived Neural Crest Model Unveils CD55 as a Cancer Stem Cell Regulator for Therapeutic Targeting in MYCN-Amplified Neuroblastoma. Neuro Oncol. 2022, 24, 872–885. [Google Scholar] [CrossRef] [PubMed]
  169. Ikeda, J.I.; Morii, E.; Liu, Y.; Qiu, Y.; Nakamichi, N.; Jokoji, R.; Miyoshi, Y.; Noguchi, S.; Aozasa, K. Prognostic Significance of CD55 Expression in Breast Cancer. Clin. Cancer Res. 2008, 14, 4780–4786. [Google Scholar] [CrossRef] [PubMed]
  170. Saygin, C.; Wiechert, A.; Rao, V.S.; Alluri, R.; Connor, E.; Thiagarajan, P.S.; Hale, J.S.; Li, Y.; Chumakova, A.; Jarrar, A.; et al. CD55 Regulates Self-Renewal and Cisplatin Resistance in Endometrioid Tumors. J. Exp. Med. 2017, 214, 2715–2732. [Google Scholar] [CrossRef]
  171. Leung, T.H.Y.; Tang, H.W.M.; Siu, M.K.Y.; Chan, D.W.; Chan, K.K.L.; Cheung, A.N.Y.; Ngan, H.Y.S. Human Papillomavirus E6 Protein Enriches the CD55(+) Population in Cervical Cancer Cells, Promoting Radioresistance and Cancer Aggressiveness. J. Pathol. 2018, 244, 151–163. [Google Scholar] [CrossRef] [PubMed]
  172. Dho, S.H.; Cho, E.H.; Lee, J.Y.; Lee, S.; Jung, S.H.; Kim, L.K.; Lim, J.C. A Novel Therapeutic Anti CD55 Monoclonal Antibody Inhibits the Proliferation and Metastasis of Colorectal Cancer Cells. Available online: https://www.spandidos-publications.com/10.3892/or.2019.7337 (accessed on 16 January 2024).
  173. Macor, P.; Tripodo, C.; Zorzet, S.; Piovan, E.; Bossi, F.; Marzari, R.; Amadori, A.; Tedesco, F. In Vivo Targeting of Human Neutralizing Antibodies against CD55 and CD59 to Lymphoma Cells Increases the Antitumor Activity of Rituximab. Cancer Res. 2007, 67, 10556–10563. [Google Scholar] [CrossRef]
  174. Dho, S.H.; Kim, S.Y.; Chung, C.; Cho, E.H.; Lee, S.Y.; Kim, J.Y.; Kim, L.K.; Min, S.W.; Lee, J.; Jung, S.H.; et al. Development of a Radionuclide-Labeled Monoclonal Anti-CD55 Antibody with Theranostic Potential in Pleural Metastatic Lung Cancer. Sci. Rep. 2018, 8, 8960. [Google Scholar] [CrossRef]
  175. Chaudhary, A.K.; Shi, J.; Cai, W.; Wang, B.; Zaman, M.S.; Huang, C.; Lin, J.; Kan, S.Z.; Zhou, J.; Dong, J.; et al. Abstract LB067: Development of a Novel Bispecific Antibody Targeting PD-L1 and CD55 for Cancer Therapy. Cancer Res. 2021, 81 (Suppl. S13), LB067. [Google Scholar] [CrossRef]
  176. Ullenhag, G.J.; Spendlove, I.; Watson, N.F.S.; Indar, A.A.; Dube, M.; Robins, R.A.; Maxwell-Armstrong, C.; Scholefield, J.H.; Durrant, L.G. A Neoadjuvant/Adjuvant Randomized Trial of Colorectal Cancer Patients Vaccinated with an Anti-Idiotypic Antibody, 105AD7, Mimicking CD55. Clin. Cancer Res. 2006, 12, 7389–7396. [Google Scholar] [CrossRef]
  177. Pritchard-Jones, K.; Spendlove, I.; Wilton, C.; Whelan, J.; Weeden, S.; Lewis, I.; Hale, J.; Douglas, C.; Pagonis, C.; Campbell, B.; et al. Immune Responses to the 105AD7 Human Anti-Idiotypic Vaccine after Intensive Chemotherapy, for Osteosarcoma. Br. J. Cancer 2005, 92, 1358. [Google Scholar] [CrossRef]
  178. Denton, G.W.L.; Durrant, L.G.; Hardcastle, J.D.; Austin, E.B.; Sewell, H.F.; Robins, R.A. Clinical Outcome of Colorectal Cancer Patients Treated with Human Monoclonal Anti-Idiotypic Antibody. Int. J. Cancer 1994, 57, 10–14. [Google Scholar] [CrossRef] [PubMed]
  179. Maxwell-Armstrong, C.A.; Durrant, L.G.; Buckley, T.J.D.; Scholefield, J.H.; Robins, R.A.; Fielding, K.; Monson, J.R.T.; Guillou, P.; Calvert, H.; Carmichael, J.; et al. Randomized Double-Blind Phase II Survival Study Comparing Immunization with the Anti-Idiotypic Monoclonal Antibody 105AD7 against Placebo in Advanced Colorectal Cancer. Br. J. Cancer 2001, 84, 1443–1446. [Google Scholar] [CrossRef]
  180. Ten-Year Follow-Up of a Prospective Trial for the Targeted Therapy of Gastric Cancer with the Human Monoclonal Antibody PAT-SC1. Available online: https://www.spandidos-publications.com/or/31/3/1059 (accessed on 19 January 2024).
  181. Liu, G.; Yin, Q.; Ji, H.; Wang, Y.; Liu, H.; Jiang, L.; Zhu, F.; Li, B. A Study on Screening and Antitumor Effect of CD55-Specific Ligand Peptide in Cervical Cancer Cells. Drug Des. Devel Ther. 2018, 12, 3899–3912. [Google Scholar] [CrossRef]
  182. Liu, G.; Xu, X.; Jiang, L.; Ji, H.; Zhu, F.; Jin, B.; Han, J.; Dong, X.; Yang, F.; Li, B. Targeted Antitumor Mechanism of C-PC/CMC-CD55sp Nanospheres in HeLa Cervical Cancer Cells. Front. Pharmacol. 2020, 11, 906. [Google Scholar] [CrossRef] [PubMed]
  183. Holla, V.R.; Wang, D.; Brown, J.R.; Mann, J.R.; Katkuri, S.; Dubois, R.N. Prostaglandin E 2 Regulates the Complement Inhibitor CD55/Decay-Accelerating Factor in Colorectal Cancer. J. Biol. Chem. 2005, 280, 476–483. [Google Scholar] [CrossRef]
  184. Oyama, S.; Fujino, H.; Yamazaki, R.; Okura, I.; Regan, J.W.; Awata, A.; Arai, T.; Murayama, T. A Novel Indole Compound, AWT-489, Inhibits Prostaglandin D2-Induced CD55 Expression by Acting on DP Prostanoid Receptors as an Antagonist in LS174T Human Colon Cancer Cells. Arch. Biochem. Biophys. 2014, 541, 21–29. [Google Scholar] [CrossRef] [PubMed]
  185. Mamidi, S.; Cinci, M.; Hasmann, M.; Fehring, V.; Kirschfink, M. Lipoplex Mediated Silencing of Membrane Regulators (CD46, CD55 and CD59) Enhances Complement-Dependent Anti-Tumor Activity of Trastuzumab and Pertuzumab. Mol. Oncol. 2013, 7, 580–594. [Google Scholar] [CrossRef]
  186. Pawig, L.; Klasen, C.; Weber, C.; Bernhagen, J.; Noels, H. Diversity and Inter-Connections in the CXCR4 Chemokine Receptor/Ligand Family: Molecular Perspectives. Front. Immunol. 2015, 6, 156039. [Google Scholar] [CrossRef]
  187. Schiraldi, M.; Raucci, A.; Muñoz, L.M.; Livoti, E.; Celona, B.; Venereau, E.; Apuzzo, T.; De Marchis, F.; Pedotti, M.; Bachi, A.; et al. HMGB1 Promotes Recruitment of Inflammatory Cells to Damaged Tissues by Forming a Complex with CXCL12 and Signaling via CXCR4. J. Exp. Med. 2012, 209, 551–563. [Google Scholar] [CrossRef] [PubMed]
  188. Chatterjee, S.; Behnam Azad, B.; Nimmagadda, S. The Intricate Role of CXCR4 in Cancer. Adv. Cancer Res. 2014, 124, 31. [Google Scholar] [CrossRef] [PubMed]
  189. Kawaguchi, N.; Zhang, T.T.; Nakanishi, T. Involvement of CXCR4 in Normal and Abnormal Development. Cells 2019, 8, 185. [Google Scholar] [CrossRef] [PubMed]
  190. Zhou, J.; Le, K.; Xu, M.; Ming, J.; Yang, W.; Zhang, Q.; Lu, L.; Xi, Z.; Ruan, S.; Huang, T. CXCR4 Antagonist AMD3100 Reverses the Resistance to Tamoxifen in Breast Cancer via Inhibiting AKT Phosphorylation. Mol. Ther. Oncolytics 2020, 18, 161–170. [Google Scholar] [CrossRef] [PubMed]
  191. Guo, F.; Wang, Y.; Liu, J.; Mok, S.C.; Xue, F.; Zhang, W. CXCL12/CXCR4: A Symbiotic Bridge Linking Cancer Cells and Their Stromal Neighbors in Oncogenic Communication Networks. Oncogene 2016, 35, 816–826. [Google Scholar] [CrossRef] [PubMed]
  192. Zhao, H.; Jiang, R.; Zhang, C.; Feng, Z.; Wang, X. The Regulatory Role of Cancer Stem Cell Marker Gene CXCR4 in the Growth and Metastasis of Gastric Cancer. npj Precis. Oncol. 2023, 7, 86. [Google Scholar] [CrossRef] [PubMed]
  193. Fujita, T.; Chiwaki, F.; Takahashi, R.U.; Aoyagi, K.; Yanagihara, K.; Nishimura, T.; Tamaoki, M.; Komatsu, M.; Komatsuzaki, R.; Matsusaki, K.; et al. Identification and Characterization of CXCR4-Positive Gastric Cancer Stem Cells. PLoS ONE 2015, 10, e0130808. [Google Scholar] [CrossRef]
  194. Dubrovska, A.; Elliott, J.; Salamone, R.J.; Telegeev, G.D.; Stakhovsky, A.E.; Schepotin, I.B.; Yan, F.; Wang, Y.; Bouchez, L.C.; Kularatne, S.A.; et al. CXCR4 Expression in Prostate Cancer Progenitor Cells. PLoS ONE 2012, 7, e31226. [Google Scholar] [CrossRef] [PubMed]
  195. Cioffi, M.; Dalterio, C.; Camerlingo, R.; Tirino, V.; Consales, C.; Riccio, A.; Ieranò, C.; Cecere, S.C.; Losito, N.S.; Greggi, S.; et al. Identification of a Distinct Population of CD133+CXCR4+ Cancer Stem Cells in Ovarian Cancer. Sci. Rep. 2015, 5, 10357. [Google Scholar] [CrossRef]
  196. Dubrovska, A.; Hartung, A.; Bouchez, L.C.; Walker, J.R.; Reddy, V.A.; Cho, C.Y.; Schultz, P.G. CXCR4 Activation Maintains a Stem Cell Population in Tamoxifen-Resistant Breast Cancer Cells through AhR Signaling. Br. J. Cancer 2012, 107, 43–52. [Google Scholar] [CrossRef]
  197. Crees, Z.D.; Rettig, M.P.; Jayasinghe, R.G.; Stockerl-Goldstein, K.; Larson, S.M.; Arpad, I.; Milone, G.A.; Martino, M.; Stiff, P.; Sborov, D.; et al. Motixafortide and G-CSF to Mobilize Hematopoietic Stem Cells for Autologous Transplantation in Multiple Myeloma: A Randomized Phase 3 Trial. Nat. Med. 2023, 29, 869–879. [Google Scholar] [CrossRef]
  198. Kuhne, M.R.; Mulvey, T.; Belanger, B.; Chen, S.; Pan, C.; Chong, C.; Cao, F.; Niekro, W.; Kempe, T.; Henning, K.A.; et al. BMS-936564/MDX-1338: A Fully Human Anti-CXCR4 Antibody Induces Apoptosis in Vitro and Shows Antitumor Activity in Vivo in Hematologic Malignancies. Clin. Cancer Res. 2013, 19, 357–366. [Google Scholar] [CrossRef] [PubMed]
  199. Kashyap, M.K.; Kumar, D.; Jones, H.; Amaya-Chanaga, C.I.; Choi, M.Y.; Melo-Cardenas, J.; Ale-Ali, A.; Kuhne, M.R.; Sabbatini, P.; Cohen, L.J.; et al. Ulocuplumab (BMS-936564/MDX1338): A Fully Human Anti-CXCR4 Antibody Induces Cell Death in Chronic Lymphocytic Leukemia Mediated through a Reactive Oxygen Species-Dependent Pathway. Oncotarget 2016, 7, 2809–2822. [Google Scholar] [CrossRef]
  200. Azad, B.B.; Chatterjee, S.; Lesniak, W.G.; Lisok, A.; Pullambhatla, M.; Bhujwalla, Z.M.; Pomper, M.G.; Nimmagadda, S. A Fully Human CXCR4 Antibody Demonstrates Diagnostic Utility and Therapeutic Efficacy in Solid Tumor Xenografts. Oncotarget 2016, 7, 12344. [Google Scholar] [CrossRef]
  201. Roccaro, A.M.; Mishima, Y.; Sacco, A.; Moschetta, M.; Tai, Y.T.; Shi, J.; Zhang, Y.; Reagan, M.R.; Huynh, D.; Kawano, Y.; et al. CXCR4 Regulates Extra-Medullary Myeloma through Epithelial-Mesenchymal-Transition-like Transcriptional Activation. Cell Rep. 2015, 12, 622–635. [Google Scholar] [CrossRef] [PubMed]
  202. Treon, S.P.; Meid, K.; Hunter, Z.R.; Flynn, C.A.; Sarosiek, S.R.; Leventoff, C.R.; White, T.P.; Cao, Y.; Roccaro, A.M.; Sacco, A.; et al. Phase 1 Study of Ibrutinib and the CXCR4 Antagonist Ulocuplumab in CXCR4-Mutated Waldenström Macroglobulinemia. Blood 2021, 138, 1535–1539. [Google Scholar] [CrossRef] [PubMed]
  203. Ghobrial, I.M.; Liu, C.J.; Redd, R.A.; Perez, R.P.; Baz, R.; Zavidij, O.; Sklavenitis-Pistofidis, R.; Richardson, P.G.; Anderson, K.C.; Laubach, J.; et al. A Phase Ib/II Trial of the First-in-Class Anti-CXCR4 Antibody Ulocuplumab in Combination with Lenalidomide or Bortezomib Plus Dexamethasone in Relapsed Multiple Myeloma. Clin. Cancer Res. 2020, 26, 344–353. [Google Scholar] [CrossRef] [PubMed]
  204. Liu, S.H.; Gu, Y.; Pascual, B.; Yan, Z.; Hallin, M.; Zhang, C.; Fan, C.; Wang, W.; Lam, J.; Spilker, M.E.; et al. A Novel CXCR4 Antagonist IgG1 Antibody (PF-06747143) for the Treatment of Hematologic Malignancies. Blood Adv. 2017, 1, 1088–1100. [Google Scholar] [CrossRef] [PubMed]
  205. Zhang, Y.; Saavedra, E.; Tang, R.; Gu, Y.; Lappin, P.; Trajkovic, D.; Liu, S.H.; Smeal, T.; Fantin, V.; De Botton, S.; et al. Targeting Primary Acute Myeloid Leukemia with a New CXCR4 Antagonist IgG1 Antibody (PF-06747143). Sci. Rep. 2017, 7, 7305. [Google Scholar] [CrossRef]
  206. Kashyap, M.K.; Amaya-Chanaga, C.I.; Kumar, D.; Simmons, B.; Huser, N.; Gu, Y.; Hallin, M.; Lindquist, K.; Yafawi, R.; Choi, M.Y.; et al. Targeting the CXCR4 Pathway Using a Novel Anti-CXCR4 IgG1 Antibody (PF-06747143) in Chronic Lymphocytic Leukemia. J. Hematol. Oncol. 2017, 10, 112. [Google Scholar] [CrossRef]
  207. Gelmini, S.; Mangoni, M.; Castiglione, F.; Beltrami, C.; Pieralli, A.; Andersson, K.L.; Fambrini, M.; Taddei, G.L.; Serio, M.; Orlando, C. The CXCR4/CXCL12 Axis in Endometrial Cancer. Clin. Exp. Metastasis 2009, 26, 261–268. [Google Scholar] [CrossRef] [PubMed]
  208. Brennecke, P.; Arlt, M.J.E.; Campanile, C.; Husmann, K.; Gvozdenovic, A.; Apuzzo, T.; Thelen, M.; Born, W.; Fuchs, B. CXCR4 Antibody Treatment Suppresses Metastatic Spread to the Lung of Intratibial Human Osteosarcoma Xenografts in Mice. Clin. Exp. Metastasis 2014, 31, 339–349. [Google Scholar] [CrossRef] [PubMed]
  209. De Pauw, T.; De Mey, L.; Debacker, J.M.; Raes, G.; Van Ginderachter, J.A.; De Groof, T.W.M.; Devoogdt, N. Current Status and Future Expectations of Nanobodies in Oncology Trials. Expert Opin. Investig. Drugs 2023, 32, 705–721. [Google Scholar] [CrossRef] [PubMed]
  210. Bin Peng, S.; Zhang, X.; Paul, D.; Kays, L.M.; Ye, M.; Vaillancourt, P.; Dowless, M.; Stancato, L.F.; Stewart, J.; Uhlik, M.T.; et al. Inhibition of CXCR4 by LY2624587, a Fully Humanized Anti-CXCR4 Antibody Induces Apoptosis of Hematologic Malignancies. PLoS ONE 2016, 11, e0150585. [Google Scholar] [CrossRef]
  211. Broussas, M.; Boute, N.; Akla, B.; Berger, S.; Beau-Larvor, C.; Champion, T.; Robert, A.; Beck, A.; Haeuw, J.F.; Goetsch, L.; et al. A New Anti-CXCR4 Antibody That Blocks the CXCR4/SDF-1 Axis and Mobilizes Effector Cells. Mol. Cancer Ther. 2016, 15, 1890–1899. [Google Scholar] [CrossRef] [PubMed]
  212. Kamal, A.; Wang, Y.; Steiner, P.; Mazzola, A.-M.; Wetzel, L.; Passino, M.; McDermott, B.; Huang, K.; Bedian, V.; Greenberg, N. Abstract 5462: MEDI3185, a Potent Anti-CXCR4 Antibody, Inhibits Tumor Cell Migration, Signaling and Tumor Growth in Preclinical Models. Cancer Res. 2013, 73 (Suppl. S8), 5462. [Google Scholar] [CrossRef]
  213. Fouquet, G.; Guidez, S.; Richez, V.; Stoppa, A.-M.; Le Tourneau, C.; Macro, M.; Gruchet, C.; Bobin, A.; Moya, N.; Systchenko, T.; et al. Phase I Dose-Escalation Study of F50067, a Humanized Anti-CXCR4 Monoclonal Antibody Alone and in Combination with Lenalidomide and Low-Dose Dexamethasone, in Relapsed or Refractory Multiple Myeloma. Oncotarget 2018, 9, 23890–23899. [Google Scholar] [CrossRef] [PubMed]
  214. Chaudary, N.; Pintilie, M.; Jelveh, S.; Lindsay, P.; Hill, R.P.; Milosevic, M. Plerixafor Improves Primary Tumor Response and Reduces Metastases in Cervical Cancer Treated with Radio-Chemotherapy. Clin. Cancer Res. 2017, 23, 1242–1249. [Google Scholar] [CrossRef] [PubMed]
  215. Andritsos, L.A.; Byrd, J.C.; Cheverton, P.; Wu, J.; Sivina, M.; Kipps, T.J.; Burger, J.A. A Multicenter Phase 1 Study of Plerixafor and Rituximab in Patients with Chronic Lymphocytic Leukemia. Leuk. Lymphoma 2019, 60, 3461–3469. [Google Scholar] [CrossRef]
  216. Heckmann, D.; Maier, P.; Laufs, S.; Wenz, F.; Zeller, W.J.; Fruehauf, S.; Allgayer, H. CXCR4 Expression and Treatment with SDF-1α or Plerixafor Modulate Proliferation and Chemosensitivity of Colon Cancer Cells. Transl. Oncol. 2013, 6, 124. [Google Scholar] [CrossRef]
  217. Taromi, S.; Kayser, G.; Catusse, J.; von Elverfeldt, D.; Reichardt, W.; Braun, F.; Weber, W.A.; Zeiser, R.; Burger, M. CXCR4 Antagonists Suppress Small Cell Lung Cancer Progression. Oncotarget 2016, 7, 85185. [Google Scholar] [CrossRef] [PubMed]
  218. Lefort, S.; Thuleau, A.; Kieffer, Y.; Sirven, P.; Bieche, I.; Marangoni, E.; Vincent-Salomon, A.; Mechta-Grigoriou, F. CXCR4 Inhibitors Could Benefit to HER2 but Not to Triple-Negative Breast Cancer Patients. Oncogene 2017, 36, 1211–1222. [Google Scholar] [CrossRef] [PubMed]
  219. Saur, D.; Seidler, B.; Schneider, G.; Algül, H.; Beck, R.; Senekowitsch-Schmidtke, R.; Schwaiger, M.; Schmid, R.M. CXCR4 Expression Increases Liver and Lung Metastasis in a Mouse Model of Pancreatic Cancer. Gastroenterology 2005, 129, 1237–1250. [Google Scholar] [CrossRef]
  220. Rubin, J.B.; Kung, A.L.; Klein, R.S.; Chan, J.A.; Sun, Y.P.; Schmidt, K.; Kieran, M.W.; Luster, A.D.; Segal, R.A. A Small-Molecule Antagonist of CXCR4 Inhibits Intracranial Growth of Primary Brain Tumors. Proc. Natl. Acad. Sci. USA 2003, 100, 13513–13518. [Google Scholar] [CrossRef] [PubMed]
  221. Domanska, U.M.; Timmer-Bosscha, H.; Nagengast, W.B.; Oude Munnink, T.H.; Kruizinga, R.C.; Ananias, H.J.K.; Kliphuis, N.M.; Huls, G.; De Vries, E.G.E.; De Jong, I.J.; et al. CXCR4 Inhibition with AMD3100 Sensitizes Prostate Cancer to Docetaxel Chemotherapy. Neoplasia 2012, 14, 709. [Google Scholar] [CrossRef]
  222. Chen, I.X.; Chauhan, V.P.; Posada, J.; Ng, M.R.; Wu, M.W.; Adstamongkonkul, P.; Huang, P.; Lindeman, N.; Langer, R.; Jain, R.K. Blocking CXCR4 Alleviates Desmoplasia, Increases T-Lymphocyte Infiltration, and Improves Immunotherapy in Metastatic Breast Cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 4558–4566. [Google Scholar] [CrossRef]
  223. Choueiri, T.K.; Atkins, M.B.; Rose, T.L.; Alter, R.S.; Ju, Y.; Niland, K.; Wang, Y.; Arbeit, R.; Parasuraman, S.; Gan, L.; et al. A Phase 1b Trial of the CXCR4 Inhibitor Mavorixafor and Nivolumab in Advanced Renal Cell Carcinoma Patients with No Prior Response to Nivolumab Monotherapy. Investig. New Drugs 2021, 39, 1019–1027. [Google Scholar] [CrossRef] [PubMed]
  224. Andtbacka, R.H.I.; Wang, Y.; Pierce, R.H.; Campbell, J.S.; Yushak, M.; Milhem, M.; Ross, M.; Niland, K.; Arbeit, R.D.; Parasuraman, S.; et al. Mavorixafor, an Orally Bioavailable CXCR4 Antagonist, Increases Immune Cell Infiltration and Inflammatory Status of Tumor Microenvironment in Patients with Melanoma. Cancer Res. Commun. 2022, 2, 904–913. [Google Scholar] [CrossRef]
  225. Gravina, G.L.; Mancini, A.; Marampon, F.; Colapietro, A.; Delle Monache, S.; Sferra, R.; Vitale, F.; Richardson, P.J.; Patient, L.; Burbidge, S.; et al. The Brain-Penetrating CXCR4 Antagonist, PRX177561, Increases the Antitumor Effects of Bevacizumab and Sunitinib in Preclinical Models of Human Glioblastoma. J. Hematol. Oncol. 2017, 10, 5. [Google Scholar] [CrossRef]
  226. Gravina, G.L.; Mancini, A.; Colapietro, A.; Vitale, F.; Vetuschi, A.; Pompili, S.; Rossi, G.; Marampon, F.; Richardson, P.J.; Patient, L.; et al. The Novel CXCR4 Antagonist, PRX177561, Reduces Tumor Cell Proliferation and Accelerates Cancer Stem Cell Differentiation in Glioblastoma Preclinical Models. Tumor Biol. 2017, 39, 1010428317695528. [Google Scholar] [CrossRef]
  227. Liang, Z.; Zhan, W.; Zhu, A.; Yoon, Y.; Lin, S.; Sasaki, M.; Klapproth, J.M.A.; Yang, H.; Grossniklaus, H.E.; Xu, J.; et al. Development of a Unique Small Molecule Modulator of CXCR4. PLoS ONE 2012, 7, e34038. [Google Scholar] [CrossRef] [PubMed]
  228. Rebolledo-Bustillo, M.; Garcia-Gomez, D.; Dávila, E.M.; Castro, M.E.; Caballero, N.A.; Melendez, F.J.; Baizabal-Aguirre, V.M.; Sanchez-Gaytan, B.L.; Perez-Aguilar, J.M. Structural Basis of the Binding Mode of the Antineoplastic Compound Motixafortide (BL-8040) in the CXCR4 Chemokine Receptor. Int. J. Mol. Sci. 2023, 24, 4393. [Google Scholar] [CrossRef] [PubMed]
  229. Borthakur, G.; Ofran, Y.; Tallman, M.S.; Foran, J.; Uy, G.L.; DiPersio, J.F.; Showel, M.M.; Shimoni, A.; Nagler, A.; Rowe, J.M.; et al. BL-8040 CXCR4 Antagonist Is Safe and Demonstrates Antileukemic Activity in Combination with Cytarabine for the Treatment of Relapsed/Refractory Acute Myelogenous Leukemia: An Open-Label Safety and Efficacy Phase 2a Study. Cancer 2021, 127, 1246–1259. [Google Scholar] [CrossRef] [PubMed]
  230. Abraham, M.; Klein, S.; Bulvik, B.; Wald, H.; Weiss, I.D.; Olam, D.; Weiss, L.; Beider, K.; Eizenberg, O.; Wald, O.; et al. The CXCR4 Inhibitor BL-8040 Induces the Apoptosis of AML Blasts by Downregulating ERK, BCL-2, MCL-1 and Cyclin-D1 via Altered MiR-15a/16-1 Expression. Leukemia 2017, 31, 2336–2346. [Google Scholar] [CrossRef]
  231. Bockorny, B.; Semenisty, V.; Macarulla, T.; Borazanci, E.; Wolpin, B.M.; Stemmer, S.M.; Golan, T.; Geva, R.; Borad, M.J.; Pedersen, K.S.; et al. BL-8040, a CXCR4 Antagonist, in Combination with Pembrolizumab and Chemotherapy for Pancreatic Cancer: The COMBAT Trial. Nat. Med. 2020, 26, 878–885. [Google Scholar] [CrossRef] [PubMed]
  232. Bockorny, B.; Macarulla, T.; Semenisty, V.; Borazanci, E.; Feliu, J.; Ponz-Sarvise, M.; Abad, D.G.; Oberstein, P.; Alistar, A.; Muñoz, A.; et al. Motixafortide and Pembrolizumab Combined to Nanoliposomal Irinotecan, Fluorouracil, and Folinic Acid in Metastatic Pancreatic Cancer: The COMBAT/KEYNOTE-202 Trial. Clin. Cancer Res. 2021, 27, 5020–5027. [Google Scholar] [CrossRef] [PubMed]
  233. Galsky, M.D.; Vogelzang, N.J.; Conkling, P.; Polzer, J.; Roberson, S.; Stille, J.R.; Saleh, M.; Thornton, D. A Phase I Trial of LY2510924, a CXCR4 Peptide Antagonist, in Patients with Advanced Cancer. Clin. Cancer Res. 2014, 20, 3581–3588. [Google Scholar] [CrossRef] [PubMed]
  234. Boddu, P.; Borthakur, G.; Koneru, M.; Huang, X.; Naqvi, K.; Wierda, W.; Bose, P.; Jabbour, E.; Estrov, Z.; Burger, J.; et al. Initial Report of a Phase I Study of LY2510924, Idarubicin, and Cytarabine in Relapsed/Refractory Acute Myeloid Leukemia. Front. Oncol. 2018, 8, 369. [Google Scholar] [CrossRef] [PubMed]
  235. O’Hara, M.H.; Messersmith, W.; Kindler, H.; Zhang, W.; Pitou, C.; Szpurka, A.M.; Wang, D.; Peng, S.-B.; Vangerow, B.; Khan, A.A.; et al. Safety and Pharmacokinetics of CXCR4 Peptide Antagonist, LY2510924, in Combination with Durvalumab in Advanced Refractory Solid Tumors. J. Pancreat. Cancer 2020, 6, 21. [Google Scholar] [CrossRef]
  236. Huang, E.H.; Singh, B.; Cristofanilli, M.; Gelovani, J.; Wei, C.; Vincent, L.; Cook, K.R.; Lucci, A. A CXCR4 Antagonist CTCE-9908 Inhibits Primary Tumor Growth and Metastasis of Breast Cancer. J. Surg. Res. 2009, 155, 231–236. [Google Scholar] [CrossRef]
  237. Phase I/II Study of CTCE-9908, a Novel Anticancer Agent that Inhibits CXCR4, in Patients with Advanced Solid Cancers|Molecular Cancer Therapeutics|American Association for Cancer Research. Available online: https://aacrjournals.org/mct/article/6/11_Supplement/A153/240072/Phase-I-II-study-of-CTCE-9908-a-novel-anticancer (accessed on 26 February 2024).
  238. Hamshaw, I.; Cominetti, M.M.D.; Lai, W.Y.; Searcey, M.; Mueller, A. The Development of Potent, Competitive CXCR4 Antagonists for the Prevention of Cancer Metastasis. Biochem. Pharmacol. 2023, 218, 115921. [Google Scholar] [CrossRef] [PubMed]
  239. Barzegar Behrooz, A.; Syahir, A.; Ahmad, S. CD133: Beyond a Cancer Stem Cell Biomarker. J. Drug Target. 2019, 27, 257–269. [Google Scholar] [CrossRef] [PubMed]
  240. Glumac, P.M.; LeBeau, A.M. The Role of CD133 in Cancer: A Concise Review. Clin. Transl. Med. 2018, 7, 18. [Google Scholar] [CrossRef] [PubMed]
  241. Bauer, N.; Fonseca, A.V.; Florek, M.; Freund, D.; Jászai, J.; Bornhäuser, M.; Fargeas, C.A.; Corbeil, D. New Insights into the Cell Biology of Hematopoietic Progenitors by Studying Prominin-1 (CD133). Cells Tissues Organs 2008, 188, 127–138. [Google Scholar] [CrossRef] [PubMed]
  242. Zacchigna, S.; Oh, H.; Wilsch-Bräuninger, M.; Missol-Kolka, E.; Jászai, J.; Jansen, S.; Tanimoto, N.; Tonagel, F.; Seeliger, M.; Huttner, W.B.; et al. Loss of the Cholesterol-Binding Protein Prominin-1/CD133 Causes Disk Dysmorphogenesis and Photoreceptor Degeneration. J. Neurosci. 2009, 29, 2297–2308. [Google Scholar] [CrossRef] [PubMed]
  243. Hemmati, H.D.; Nakano, I.; Lazareff, J.A.; Masterman-Smith, M.; Geschwind, D.H.; Bronner-Fraser, M.; Kornblum, H.I. Cancerous Stem Cells Can Arise from Pediatric Brain Tumors. Proc. Natl. Acad. Sci. USA 2003, 100, 15178–15183. [Google Scholar] [CrossRef] [PubMed]
  244. Identification of a Cancer Stem Cell in Human Brain Tumors|Cancer Research|American Association for Cancer Research. Available online: https://aacrjournals.org/cancerres/article/63/18/5821/510370/Identification-of-a-Cancer-Stem-Cell-in-Human (accessed on 22 February 2024).
  245. Yuan, X.; Curtin, J.; Xiong, Y.; Liu, G.; Waschsmann-Hogiu, S.; Farkas, D.L.; Black, K.L.; Yu, J.S. Isolation of Cancer Stem Cells from Adult Glioblastoma Multiforme. Oncogene 2004, 23, 9392–9400. [Google Scholar] [CrossRef] [PubMed]
  246. Singh, S.K.; Hawkins, C.; Clarke, I.D.; Squire, J.A.; Bayani, J.; Hide, T.; Henkelman, R.M.; Cusimano, M.D.; Dirks, P.B. Identification of Human Brain Tumor Initiating Cells. Nature 2004, 432, 396–401. [Google Scholar] [CrossRef] [PubMed]
  247. Vander Griend, D.J.; Karthaus, W.L.; Dalrymple, S.; Meeker, A.; DeMarzo, A.M.; Isaacs, J.T. The Role of CD133 in Normal Human Prostate Stem Cells and Malignant Cancer-Initiating Cells. Cancer Res. 2008, 68, 9703–9711. [Google Scholar] [CrossRef]
  248. Ma, S.; Tang, K.H.; Chan, Y.P.; Lee, T.K.; Kwan, P.S.; Castilho, A.; Ng, I.; Man, K.; Wong, N.; To, K.F.; et al. MiR-130b Promotes CD133(+) Liver Tumor-Initiating Cell Growth and Self-Renewal via Tumor Protein 53-Induced Nuclear Protein 1. Cell Stem Cell 2010, 7, 694–707. [Google Scholar] [CrossRef]
  249. Fang, D.D.; Kim, Y.J.; Lee, C.N.; Aggarwal, S.; McKinnon, K.; Mesmer, D.; Norton, J.; Birse, C.E.; He, T.; Ruben, S.M.; et al. Expansion of CD133+ Colon Cancer Cultures Retaining Stem Cell Properties to Enable Cancer Stem Cell Target Discovery. Br. J. Cancer 2010, 102, 1265–1275. [Google Scholar] [CrossRef] [PubMed]
  250. Eramo, A.; Lotti, F.; Sette, G.; Pilozzi, E.; Biffoni, M.; Di Virgilio, A.; Conticello, C.; Ruco, L.; Peschle, C.; De Maria, R. Identification and Expansion of the Tumorigenic Lung Cancer Stem Cell Population. Cell Death Differ. 2008, 15, 504–514. [Google Scholar] [CrossRef] [PubMed]
  251. Zhang, Q.; Shi, S.; Yen, Y.; Brown, J.; Ta, J.Q.; Le, A.D. A Subpopulation of CD133+ Cancer Stem-like Cells Characterized in Human Oral Squamous Cell Carcinoma Confer Resistance to Chemotherapy. Cancer Lett. 2010, 289, 151–160. [Google Scholar] [CrossRef] [PubMed]
  252. Joseph, C.; Arshad, M.; Kurozomi, S.; Althobiti, M.; Miligy, I.M.; Al-izzi, S.; Toss, M.S.; Goh, F.Q.; Johnston, S.J.; Martin, S.G.; et al. Overexpression of the Cancer Stem Cell Marker CD133 Confers a Poor Prognosis in Invasive Breast Cancer. Breast Cancer Res. Treat. 2019, 174, 387–399. [Google Scholar] [CrossRef] [PubMed]
  253. Chen, S.; Song, X.; Chen, Z.; Li, X.; Li, M.; Liu, H.; Li, J. CD133 Expression and the Prognosis of Colorectal Cancer: A Systematic Review and Meta-Analysis. PLoS ONE 2013, 8, e56380. [Google Scholar] [CrossRef] [PubMed]
  254. da Costa, W.H.; Rocha, R.M.; da Cunha, I.W.; da Fonseca, F.P.; Guimaraes, G.C.; de Cassio Zequi, S. CD133 Immunohistochemical Expression Predicts Progression and Cancer-Related Death in Renal Cell Carcinoma. World J. Urol. 2012, 30, 553–558. [Google Scholar] [CrossRef] [PubMed]
  255. Kim, M.P.; Fleming, J.B.; Wang, H.; Abbruzzese, J.L.; Choi, W.; Kopetz, S.; McConkey, D.J.; Evans, D.B.; Gallick, G.E. ALDH Activity Selectively Defines an Enhanced Tumor-Initiating Cell Population Relative to CD133 Expression in Human Pancreatic Adenocarcinoma. PLoS ONE 2011, 6, e20636. [Google Scholar] [CrossRef] [PubMed]
  256. Mohd-Zahid, M.H.; Zulkifli, S.N.; Che Abdullah, C.A.; Lim, J.; Fakurazi, S.; Wong, K.K.; Zakaria, A.D.; Ismail, N.; Uskoković, V.; Mohamud, R.; et al. Gold Nanoparticles Conjugated with Anti-CD133 Monoclonal Antibody and 5-Fluorouracil Chemotherapeutic Agent as Nanocarriers for Cancer Cell Targeting. RSC Adv. 2021, 11, 16131. [Google Scholar] [CrossRef] [PubMed]
  257. Swaminathan, S.K.; Roger, E.; Toti, U.; Niu, L.; Ohlfest, J.R.; Panyam, J. CD133-Targeted Paclitaxel Delivery Inhibits Local Tumor Recurrence in a Mouse Model of Breast Cancer. J. Control. Release 2013, 171, 280–287. [Google Scholar] [CrossRef]
  258. Ning, S.T.; Lee, S.Y.; Wei, M.F.; Peng, C.L.; Lin, S.Y.F.; Tsai, M.H.; Lee, P.C.; Shih, Y.H.; Lin, C.Y.; Luo, T.Y.; et al. Targeting Colorectal Cancer Stem-Like Cells with Anti-CD133 Antibody-Conjugated SN-38 Nanoparticles. ACS Appl. Mater. Interfaces 2016, 8, 17793–17804. [Google Scholar] [CrossRef]
  259. Huang, J.; Li, C.; Wang, Y.; Lv, H.; Guo, Y.; Dai, H.; Wicha, M.S.; Chang, A.E.; Li, Q. Cytokine-Induced Killer (CIK) Cells Bound with Anti-CD3/Anti-CD133 Bispecific Antibodies Target CD133high Cancer Stem Cells in Vitro and in Vivo. Clin. Immunol. 2013, 149, 156–168. [Google Scholar] [CrossRef]
  260. Riegg, F.; Lutz, M.S.; Schmied, B.J.; Heitmann, J.S.; Queudeville, M.; Lang, P.; Jung, G.; Salih, H.R.; Märklin, M.A. FC-Optimized CD133 Antibody for Induction of NK Cell Reactivity against B Cell Acute Lymphoblastic Leukemia. Cancers 2021, 13, 1632. [Google Scholar] [CrossRef]
  261. Schmied, B.J.; Riegg, F.; Zekri, L.; Grosse-Hovest, L.; Bühring, H.J.; Jung, G.; Salih, H.R. An Fc-Optimized CD133 Antibody for Induction of Natural Killer Cell Reactivity against Colorectal Cancer. Cancers 2019, 11, 789. [Google Scholar] [CrossRef]
  262. Klapdor, R.; Wang, S.; Hacker, U.; Büning, H.; Morgan, M.; Dörk, T.; Hillemanns, P.; Schambach, A. Improved Killing of Ovarian Cancer Stem Cells by Combining a Novel Chimeric Antigen Receptor–Based Immunotherapy and Chemotherapy. Hum. Gene Ther. 2017, 28, 886–896. [Google Scholar] [CrossRef]
  263. Taromi, S.; Firat, E.; Simonis, A.; Braun, L.M.; Apostolova, P.; Elze, M.; Passlick, B.; Schumacher, A.; Lagies, S.; Frey, A.; et al. Enhanced AC133-Specific CAR T Cell Therapy Induces Durable Remissions in Mice with Metastatic Small Cell Lung Cancer. Cancer Lett. 2022, 538, 215697. [Google Scholar] [CrossRef]
  264. Wang, Y.; Chen, M.; Wu, Z.; Tong, C.; Dai, H.; Guo, Y.; Liu, Y.; Huang, J.; Lv, H.; Luo, C.; et al. CD133-Directed CAR T Cells for Advanced Metastasis Malignancies: A Phase I Trial. Oncoimmunology 2018, 7, e1440169. [Google Scholar] [CrossRef]
  265. Dai, H.; Tong, C.; Shi, D.; Chen, M.; Guo, Y.; Chen, D.; Han, X.; Wang, H.; Wang, Y.; Shen, P. Efficacy and Biomarker Analysis of CD133-Directed CAR T Cells in Advanced Hepatocellular Carcinoma: A Single-Arm, Open-Label, Phase II Trial. Oncoimmunology 2020, 9, 1846926. [Google Scholar] [CrossRef]
  266. Schmohl, J.U.; Gleason, M.K.; Dougherty, P.R.; Miller, J.S.; Vallera, D.A. Heterodimeric Bispecific Single Chain Variable Fragments (ScFv) Killer Engagers (BiKEs) Enhance NK-Cell Activity Against CD133+ Colorectal Cancer Cells. Target. Oncol. 2016, 11, 353–361. [Google Scholar] [CrossRef]
  267. Schmohl, J.U.; Felices, M.; Oh, F.; Lenvik, A.J.; Lebeau, A.M.; Panyam, J.; Miller, J.S.; Vallera, D.A. Engineering of Anti-CD133 Trispecific Molecule Capable of Inducing NK Expansion and Driving Antibody-Dependent Cell-Mediated Cytotoxicity. Cancer Res. Treat. Off. J. Korean Cancer Assoc. 2017, 49, 1140–1152. [Google Scholar] [CrossRef]
  268. Schmohl, J.U.; Felices, M.; Todhunter, D.; Taras, E.; Miller, J.S.; Vallera, D.A. Tetraspecific ScFv Construct Provides NK Cell Mediated ADCC and Self-Sustaining Stimuli via Insertion of IL-15 as a Cross-Linker. Oncotarget 2016, 7, 73830. [Google Scholar] [CrossRef]
  269. Rudnick, J.D.; Fink, K.L.; Landolfi, J.C.; Markert, J.; Piccioni, D.E.; Glantz, M.J.; Swanson, S.J.; Gringeri, A.; Yu, J. Immunological Targeting of CD133 in Recurrent Glioblastoma: A Multi-Center Phase I Translational and Clinical Study of Autologous CD133 Dendritic Cell Immunotherapy. J. Clin. Oncol. 2017, 35 (Suppl. S15), 2059. [Google Scholar] [CrossRef]
  270. Hsin, I.L.; Chiu, L.Y.; Ou, C.C.; Wu, W.J.; Sheu, G.T.; Ko, J.L. CD133 Inhibition via Autophagic Degradation in Pemetrexed-Resistant Lung Cancer Cells by GMI, a Fungal Immunomodulatory Protein from Ganoderma Microsporum. Br. J. Cancer 2020, 123, 449–458. [Google Scholar] [CrossRef]
  271. Waldron, N.N.; Kaufman, D.S.; Oh, S.; Inde, Z.; Hexum, M.K.; Ohlfest, J.R.; Vallera, D.A. Targeting Tumor-Initiating Cancer Cells with DCD133KDEL Shows Impressive Tumor Reductions in a Xenotransplant Model of Human Head and Neck Cancer. Mol. Cancer Ther. 2011, 10, 1829–1838. [Google Scholar] [CrossRef]
  272. Ohlfest, J.R.; Zellmer, D.M.; Panyam, J.; Swaminathan, S.K.; Oh, S.; Waldron, N.N.; Toma, S.; Vallera, D.A. Immunotoxin Targeting CD133+ Breast Carcinoma Cells. Drug Deliv. Transl. Res. 2013, 3, 195–204. [Google Scholar] [CrossRef]
  273. Skubitz, A.P.N.; Taras, E.P.; Boylan, K.L.M.; Waldron, N.N.; Oh, S.; Panoskaltsis-Mortari, A.; Vallera, D.A. Targeting CD133 in an in Vivo Ovarian Cancer Model Reduces Ovarian Cancer Progression. Gynecol. Oncol. 2013, 130, 579–587. [Google Scholar] [CrossRef]
  274. Bostad, M.; Olsen, C.E.; Peng, Q.; Berg, K.; Høgset, A.; Selbo, P.K. Light-Controlled Endosomal Escape of the Novel CD133-Targeting Immunotoxin AC133–Saporin by Photochemical Internalization—A Minimally Invasive Cancer Stem Cell-Targeting Strategy. J. Control. Release 2015, 206, 37–48. [Google Scholar] [CrossRef]
  275. Deng, Y.; Su, Q.; Mo, J.; Fu, X.; Zhang, Y.; Lin, E.H. Celecoxib Downregulates CD133 Expression through Inhibition of the Wnt Signaling Pathway in Colon Cancer Cells. Cancer Investig. 2013, 31, 97–102. [Google Scholar] [CrossRef]
  276. Jung, K.H.; Lee, J.H.; Kim, M.; Lee, E.J.; Cho, Y.S.; Lee, K.H. Celecoxib-Induced Modulation of Colon Cancer CD133 Expression Occurs through AKT Inhibition and Is Monitored by 89Zr Immuno-PET. Mol. Imaging 2022, 2022, 4906934. [Google Scholar] [CrossRef]
  277. Tsunekuni, K.; Konno, M.; Haraguchi, N.; Koseki, J.; Asai, A.; Matsuoka, K.; Kobunai, T.; Takechi, T.; Doki, Y.; Mori, M.; et al. CD44/CD133-Positive Colorectal Cancer Stem Cells Are Sensitive to Trifluridine Exposure. Sci. Rep. 2019, 9, 14861. [Google Scholar] [CrossRef]
  278. Deng, H.; Liu, H.; Yang, G.; Wang, D.; Luo, Y.; Li, C.; Qi, Z.; Liu, Z.; Wang, P.; Jia, Y.; et al. ACT001 Inhibited CD133 Transcription by Targeting and Inducing Olig2 Ubiquitination Degradation. Oncogenesis 2023, 12, 19. [Google Scholar] [CrossRef]
  279. Xi, X.; Liu, N.; Wang, Q.; Chu, Y.; Yin, Z.; Ding, Y.; Lu, Y. ACT001, a Novel PAI-1 Inhibitor, Exerts Synergistic Effects in Combination with Cisplatin by Inhibiting PI3K/AKT Pathway in Glioma. Cell Death Dis. 2019, 10, 757. [Google Scholar] [CrossRef]
  280. Lickliter, J.D.; Jennens, R.; Lemech, C.R.; Kichenadasse, G.; Cai, D.; Su, S.Y.-C. Phase 1 Dose-Escalation Study of ACT001 in Patients with Recurrent Glioblastoma and Other Advanced Solid Tumors. J. Clin. Oncol. 2021, 39 (Suppl. S15), 2037. [Google Scholar] [CrossRef]
  281. Yin, W.; Pham, C.V.; Wang, T.; Al Shamaileh, H.; Chowdhury, R.; Patel, S.; Li, Y.; Kong, L.; Hou, Y.; Zhu, Y.; et al. Inhibition of Autophagy Promotes the Elimination of Liver Cancer Stem Cells by CD133 Aptamer-Targeted Delivery of Doxorubicin. Biomolecules 2022, 12, 1623. [Google Scholar] [CrossRef]
  282. Zhou, G.; Da Won Bae, S.; Nguyen, R.; Huo, X.; Han, S.; Zhang, Z.; Hebbard, L.; Duan, W.; Eslam, M.; Liddle, C.; et al. An Aptamer-Based Drug Delivery Agent (CD133-Apt-Dox) Selectively and Effectively Kills Liver Cancer Stem-like Cells. Cancer Lett. 2021, 501, 124–132. [Google Scholar] [CrossRef]
  283. Fang, H.; Xie, J.; Zhang, M.; Zhao, Z.; Wan, Y.; Yao, Y. MiRNA-21 Promotes Proliferation and Invasion of Triple-Negative Breast Cancer Cells through Targeting PTEN. Am. J. Transl. Res. 2017, 9, 953. [Google Scholar]
  284. Yin, H.; Xiong, G.; Guo, S.; Xu, C.; Xu, R.; Guo, P.; Shu, D. Delivery of Anti-MiRNA for Triple-Negative Breast Cancer Therapy Using RNA Nanoparticles Targeting Stem Cell Marker CD133. Mol. Ther. 2019, 27, 1252–1261. [Google Scholar] [CrossRef]
  285. Sun, J.; Zhang, C.; Liu, G.; Liu, H.; Zhou, C.; Lu, Y.; Zhou, C.; Yuan, L.; Li, X. A Novel Mouse CD133 Binding-Peptide Screened by Phage Display Inhibits Cancer Cell Motility in Vitro. Clin. Exp. Metastasis 2012, 29, 185–196. [Google Scholar] [CrossRef]
  286. Li, W.; Cho, M.Y.; Lee, S.; Jang, M.; Park, J.; Park, R. CRISPR-Cas9 Mediated CD133 Knockout Inhibits Colon Cancer Invasion through Reduced Epithelial-Mesenchymal Transition. PLoS ONE 2019, 14, e0220860. [Google Scholar] [CrossRef]
  287. Asadzadeh, Z.; Mansoori, B.; Mohammadi, A.; Kazemi, T.; Mokhtarzadeh, A.; Shanehbandi, D.; Hemmat, N.; Derakhshani, A.; Brunetti, O.; Safaei, S.; et al. The Combination Effect of Prominin1 (CD133) Suppression and Oxaliplatin Treatment in Colorectal Cancer Therapy. Biomed. Pharmacother. 2021, 137, 111364. [Google Scholar] [CrossRef]
  288. Jing, H.; Weidensteiner, C.; Reichardt, W.; Gaedicke, S.; Zhu, X.; Grosu, A.L.; Kobayashi, H.; Niedermann, G. Imaging and Selective Elimination of Glioblastoma Stem Cells with Theranostic Near-Infrared-Labeled CD133-Specific Antibodies. Theranostics 2016, 6, 862. [Google Scholar] [CrossRef]
  289. Cavaleri, F.; Schöler, H.R. Nanog: A New Recruit to the Embryonic Stem Cell Orchestra. Cell 2003, 113, 551–552. [Google Scholar] [CrossRef]
  290. Jeter, C.R.; Liu, B.; Liu, X.; Chen, X.; Liu, C.; Calhoun-Davis, T.; Repass, J.; Zaehres, H.; Shen, J.J.; Tang, D.G. NANOG Promotes Cancer Stem Cell Characteristics and Prostate Cancer Resistance to Androgen Deprivation. Oncogene 2011, 30, 3833–3845. [Google Scholar] [CrossRef]
  291. Rodrigo, J.P.; Villaronga, M.Á.; Menéndez, S.T.; Hermida-Prado, F.; Quer, M.; Vilaseca, I.; Allonca, E.; Pedregal Mallo, D.; Astudillo, A.; García-Pedrero, J.M. A Novel Role for Nanog As An Early Cancer Risk Marker In Patients with Laryngeal Precancerous Lesions. Sci. Rep. 2017, 7, 11110. [Google Scholar] [CrossRef]
  292. Lu, X.; Mazur, S.J.; Lin, T.; Appella, E.; Xu, Y. The Pluripotency Factor Nanog Promotes Breast Cancer Tumorigenesis and Metastasis. Oncogene 2013, 33, 2655–2664. [Google Scholar] [CrossRef]
  293. Vasefifar, P.; Motafakkerazad, R.; Maleki, L.A.; Najafi, S.; Ghrobaninezhad, F.; Najafzadeh, B.; Alemohammad, H.; Amini, M.; Baghbanzadeh, A.; Baradaran, B. Nanog, as a Key Cancer Stem Cell Marker in Tumor Progression. Gene 2022, 827, 146448. [Google Scholar] [CrossRef]
  294. Begicevic, R.R.; Falasca, M. ABC Transporters in Cancer Stem Cells: Beyond Chemoresistance. Int. J. Mol. Sci. 2017, 18, 2362. [Google Scholar] [CrossRef] [PubMed]
  295. Zhao, L.; Liu, J.; Chen, S.; Fang, C.; Zhang, X.; Luo, Z. Prognostic Significance of NANOG Expression in Solid Tumors: A Meta-Analysis. Onco Targets Ther. 2018, 11, 5515–5526. [Google Scholar] [CrossRef]
  296. Jeter, C.R.; Badeaux, M.; Choy, G.; Chandra, D.; Patrawala, L.; Liu, C.; Calhoun-Davis, T.; Zaehres, H.; Daley, G.Q.; Tang, D.G. Functional Evidence That the Self-Renewal Gene NANOG Regulates Human Tumor Development. Stem Cells 2009, 27, 993–1005. [Google Scholar] [CrossRef]
  297. Ibrahim, E.E.; Babaei-Jadidi, R.; Saadeddin, A.; Spencer-Dene, B.; Hossaini, S.; Abuzinadah, M.; Li, N.; Fadhil, W.; Ilyas, M.; Bonnet, D.; et al. Embryonic NANOG Activity Defines Colorectal Cancer Stem Cells and Modulates through AP1- and TCF-Dependent Mechanisms. Stem Cells 2012, 30, 2076–2087. [Google Scholar] [CrossRef]
  298. Shan, J.; Shen, J.; Liu, L.; Xia, F.; Xu, C.; Duan, G.; Xu, Y.; Ma, Q.; Yang, Z.; Zhang, Q.; et al. Nanog Regulates Self-Renewal of Cancer Stem Cells through the Insulin-like Growth Factor Pathway in Human Hepatocellular Carcinoma. Hepatology 2012, 56, 1004–1014. [Google Scholar] [CrossRef]
  299. Qin, J.; Liu, X.; Laffin, B.; Chen, X.; Choy, G.; Jeter, C.R.; Calhoun-Davis, T.; Li, H.; Palapattu, G.S.; Pang, S.; et al. The PSA-/Lo Prostate Cancer Cell Population Harbors Self-Renewing Long-Term Tumor-Propagating Cells That Resist Castration. Cell Stem Cell 2012, 10, 556–569. [Google Scholar] [CrossRef]
  300. Liu, Y.; Clem, B.; Zuba-Surma, E.K.; El-Naggar, S.; Telang, S.; Jenson, A.B.; Wang, Y.; Shao, H.; Ratajczak, M.Z.; Chesney, J.; et al. Mouse Fibroblasts Lacking RB1 Function Form Spheres and Undergo Reprogramming to a Cancer Stem Cell Phenotype. Cell Stem Cell 2009, 4, 336–347. [Google Scholar] [CrossRef]
  301. Xiang, T.; Long, H.; He, L.; Han, X.; Lin, K.; Liang, Z.; Zhuo, W.; Xie, R.; Zhu, B. Interleukin-17 Produced by Tumor Microenvironment Promotes Self-Renewal of CD133+ Cancer Stem-like Cells in Ovarian Cancer. Oncogene 2013, 34, 165–176. [Google Scholar] [CrossRef]
  302. Kundu, J.; Banerjee, P.; Bose, C.; Das, U.; Ghosh, U.; Sinha, S. Internal Oligoguanidinium Transporter: Mercury-Free Scalable Synthesis, Improvement of Cellular Localization, Endosomal Escape, Mitochondrial Localization, and Conjugation with Antisense Morpholino for NANOG Inhibition to Induce Chemosensitization of Taxol in MCF-7 Cells. Bioconjug. Chem. 2020, 31, 2367–2382. [Google Scholar] [CrossRef]
  303. Summerton, J. Morpholino Antisense Oligomers: The Case for an RNase H-Independent Structural Type. Biochim. Biophys. Acta 1999, 1489, 141–158. [Google Scholar] [CrossRef]
  304. Godfrey, C.; Desviat, L.R.; Smedsrød, B.; Piétri-Rouxel, F.; Denti, M.A.; Disterer, P.; Lorain, S.; Nogales-Gadea, G.; Sardone, V.; Anwar, R.; et al. Delivery Is Key: Lessons Learnt from Developing Splice-switching Antisense Therapies. EMBO Mol. Med. 2017, 9, 545. [Google Scholar] [CrossRef]
  305. Khosravi, N.; Shahgoli, V.K.; Amini, M.; Safaei, S.; Mokhtarzadeh, A.; Mansoori, B.; Derakhshani, A.; Baghbanzadeh, A.; Baradaran, B. Suppression of Nanog Inhibited Cell Migration and Increased the Sensitivity of Colorectal Cancer Cells to 5-Fluorouracil. Eur. J. Pharmacol. 2021, 894, 173871. [Google Scholar] [CrossRef]
  306. Baltus, G.A.; Kowalski, M.P.; Tutter, A.V.; Kadam, S. A Positive Regulatory Role for the MSin3A-HDAC Complex in Pluripotency through Nanog and Sox2. J. Biol. Chem. 2009, 284, 6998–7006. [Google Scholar] [CrossRef]
  307. Marks, P.A. Discovery and Development of SAHA as an Anticancer Agent. Oncogene 2007, 26, 1351–1356. [Google Scholar] [CrossRef]
  308. Kumar, B.; Yadav, A.; Lang, J.C.; Teknos, T.N.; Kumar, P. Suberoylanilide Hydroxamic Acid (SAHA) Reverses Chemoresistance in Head and Neck Cancer Cells by Targeting Cancer Stem Cells via the Downregulation of Nanog. Genes Cancer 2015, 6, 169. [Google Scholar] [CrossRef]
  309. Zhang, J.; Chen, M.; Zhu, Y.; Dai, X.; Dang, F.; Ren, J.; Ren, S.; Shulga, Y.V.; Beca, F.; Gan, W.; et al. SPOP Promotes Nanog Destruction to Suppress Stem Cell Traits and Prostate Cancer Progression. Dev. Cell 2019, 48, 329–344.e5. [Google Scholar] [CrossRef]
  310. Moretto-Zita, M.; Jin, H.; Shen, Z.; Zhao, T.; Briggs, S.P.; Xu, Y. Phosphorylation Stabilizes Nanog by Promoting Its Interaction with Pin1. Proc. Natl. Acad. Sci. USA 2010, 107, 13312–13317. [Google Scholar] [CrossRef] [PubMed]
  311. Uchida, T.; Takamiya, M.; Takahashi, M.; Miyashita, H.; Ikeda, H.; Terada, T.; Matsuo, Y.; Shirouzu, M.; Yokoyama, S.; Fujimori, F.; et al. Pin1 and Par14 Peptidyl Prolyl Isomerase Inhibitors Block Cell Proliferation. Chem. Biol. 2003, 10, 15–24. [Google Scholar] [CrossRef] [PubMed]
  312. Rustighi, A.; Zannini, A.; Tiberi, L.; Sommaggio, R.; Piazza, S.; Sorrentino, G.; Nuzzo, S.; Tuscano, A.; Eterno, V.; Benvenuti, F.; et al. Prolyl-Isomerase Pin1 Controls Normal and Cancer Stem Cells of the Breast. EMBO Mol. Med. 2014, 6, 99–119. [Google Scholar] [CrossRef] [PubMed]
  313. Sato, A.; Okada, M.; Shibuya, K.; Watanabe, E.; Seino, S.; Suzuki, K.; Narita, Y.; Shibui, S.; Kayama, T.; Kitanaka, C. Resveratrol Promotes Proteasome-Dependent Degradation of Nanog via P53 Activation and Induces Differentiation of Glioma Stem Cells. Stem Cell Res. 2013, 11, 601–610. [Google Scholar] [CrossRef] [PubMed]
  314. Ko, J.H.; Sethi, G.; Um, J.Y.; Shanmugam, M.K.; Arfuso, F.; Kumar, A.P.; Bishayee, A.; Ahn, K.S. The Role of Resveratrol in Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 2589. [Google Scholar] [CrossRef] [PubMed]
  315. Wang, H.; Liu, B.; Wang, J.; Li, J.; Gong, Y.; Li, S.; Wang, C.; Cui, B.; Xue, X.; Yang, M.; et al. Reduction of NANOG Mediates the Inhibitory Effect of Aspirin on Tumor Growth and Stemness in Colorectal Cancer. Cell Physiol. Biochem. 2017, 44, 1051–1063. [Google Scholar] [CrossRef] [PubMed]
  316. Shi, P.; Liu, W.; Tala, T.; Wang, H.; Li, F.; Zhang, H.; Wu, Y.; Kong, Y.; Zhou, Z.; Wang, C.; et al. Metformin Suppresses Triple-Negative Breast Cancer Stem Cells by Targeting KLF5 for Degradation. Cell Discov. 2017, 3, 17010. [Google Scholar] [CrossRef] [PubMed]
  317. Narusaka, T.; Ohara, T.; Noma, K.; Nishiwaki, N.; Katsura, Y.; Kato, T.; Sato, H.; Tomono, Y.; Kikuchi, S.; Tazawa, H.; et al. Nanog Is a Promising Chemoresistant Stemness Marker and Therapeutic Target by Iron Chelators for Esophageal Cancer. Int. J. Cancer 2021, 149, 347–357. [Google Scholar] [CrossRef] [PubMed]
  318. Zhou, B.; Lin, W.; Long, Y.; Yang, Y.; Zhang, H.; Wu, K.; Chu, Q. Notch Signaling Pathway: Architecture, Disease, and Therapeutics. Signal Transduct. Target. Ther. 2022, 7, 95. [Google Scholar] [CrossRef]
  319. Aster, J.C.; Pear, W.S.; Blacklow, S.C. The Varied Roles of Notch in Cancer. Annu. Rev. Pathol. 2017, 12, 245. [Google Scholar] [CrossRef]
  320. Kopan, R.; Ilagan, M.X.G. The Canonical Notch Signaling Pathway: Unfolding the Activation Mechanism. Cell 2009, 137, 216–233. [Google Scholar] [CrossRef]
  321. Majumder, S.; Crabtree, J.S.; Golde, T.E.; Minter, L.M.; Osborne, B.A.; Miele, L. Targeting Notch in Oncology: The Path Forward. Nat. Rev. Drug Discov. 2020, 20, 125–144. [Google Scholar] [CrossRef]
  322. Hu, Y.Y.; Zheng, M.H.; Cheng, G.; Li, L.; Liang, L.; Gao, F.; Wei, Y.N.; Fu, L.A.; Han, H. Notch Signaling Contributes to the Maintenance of Both Normal Neural Stem Cells and Patient-Derived Glioma Stem Cells. BMC Cancer 2011, 11, 82. [Google Scholar] [CrossRef]
  323. Xiao, W.; Gao, Z.; Duan, Y.; Yuan, W.; Ke, Y. Notch Signaling Plays a Crucial Role in Cancer Stem-like Cells Maintaining Stemness and Mediating Chemotaxis in Renal Cell Carcinoma. J. Exp. Clin. Cancer Res. 2017, 36, 41. [Google Scholar] [CrossRef]
  324. Abel, E.V.; Kim, E.J.; Wu, J.; Hynes, M.; Bednar, F.; Proctor, E.; Wang, L.; Dziubinski, M.L.; Simeone, D.M. The Notch Pathway Is Important in Maintaining the Cancer Stem Cell Population in Pancreatic Cancer. PLoS ONE 2014, 9, e91983. [Google Scholar] [CrossRef]
  325. D’Angelo, R.C.; Ouzounova, M.; Davis, A.; Choi, D.; Tchuenkam, S.M.; Kim, G.; Luther, T.; Quraishi, A.A.; Senbabaoglu, Y.; Conley, S.J.; et al. Notch Reporter Activity in Breast Cancer Cell Lines Identifies a Subset of Cells with Stem Cell Activity. Mol. Cancer Ther. 2015, 14, 779–787. [Google Scholar] [CrossRef] [PubMed]
  326. Allen, F.; Maillard, I. Therapeutic Targeting of Notch Signaling: From Cancer to Inflammatory Disorders. Front. Cell Dev. Biol. 2021, 9, 649205. [Google Scholar] [CrossRef]
  327. Bender, M.H.; Gao, H.; Capen, A.R.; Clay, J.M.; Hipskind, P.A.; Reel, J.K.; Zamek-Gliszczynski, M.J.; Manro, J.R.; Benhadji, K.; Patel, B.K.R. Abstract 1131: Novel Inhibitor of Notch Signaling for the Treatment of Cancer. Cancer Res. 2013, 73 (Suppl. S8), 1131. [Google Scholar] [CrossRef]
  328. Mancarella, S.; Serino, G.; Dituri, F.; Cigliano, A.; Ribback, S.; Wang, J.; Chen, X.; Calvisi, D.F.; Giannelli, G. Crenigacestat, a Selective NOTCH1 Inhibitor, Reduces Intrahepatic Cholangiocarcinoma Progression by Blocking VEGFA/DLL4/MMP13 Axis. Cell Death Differ. 2020, 27, 2330–2343. [Google Scholar] [CrossRef]
  329. Massard, C.; Azaro, A.; Soria, J.C.; Lassen, U.; Le Tourneau, C.; Sarker, D.; Smith, C.; Ohnmacht, U.; Oakley, G.; Patel, B.K.R.; et al. First-in-Human Study of LY3039478, an Oral Notch Signaling Inhibitor in Advanced or Metastatic Cancer. Ann. Oncol. 2018, 29, 1911–1917. [Google Scholar] [CrossRef]
  330. Doi, T.; Tajimi, M.; Mori, J.; Asou, H.; Inoue, K.; Benhadji, K.A.; Naito, Y. A Phase 1 Study of Crenigacestat (LY3039478), the Notch Inhibitor, in Japanese Patients with Advanced Solid Tumors. Investig. New Drugs 2021, 39, 469. [Google Scholar] [CrossRef] [PubMed]
  331. Even, C.; Lassen, U.; Merchan, J.; Le Tourneau, C.; Soria, J.C.; Ferte, C.; Ricci, F.; Diener, J.T.; Yuen, E.; Smith, C.; et al. Safety and Clinical Activity of the Notch Inhibitor, Crenigacestat (LY3039478), in an Open-Label Phase I Trial Expansion Cohort of Advanced or Metastatic Adenoid Cystic Carcinoma. Investig. New Drugs 2020, 38, 402. [Google Scholar] [CrossRef] [PubMed]
  332. Cowan, A.J.; Pont, M.J.; Sather, B.D.; Turtle, C.J.; Till, B.G.; Libby, E.N.; Coffey, D.G.; Tuazon, S.A.; Wood, B.; Gooley, T.; et al. γ-Secretase Inhibitor in Combination with BCMA Chimeric Antigen Receptor T-Cell Immunotherapy for Individuals with Relapsed or Refractory Multiple Myeloma: A Phase 1, First-in-Human Trial. Lancet Oncol. 2023, 24, 811–822. [Google Scholar] [CrossRef]
  333. Pant, S.; Jones, S.F.; Kurkjian, C.D.; Infante, J.R.; Moore, K.N.; Burris, H.A.; McMeekin, D.S.; Benhadji, K.A.; Patel, B.K.R.; Frenzel, M.J.; et al. A First-in-Human Phase I Study of the Oral Notch Inhibitor, LY900009, in Patients with Advanced Cancer. Eur. J. Cancer 2016, 56, 1–9. [Google Scholar] [CrossRef] [PubMed]
  334. Gavai, A.V.; Quesnelle, C.; Norris, D.; Han, W.C.; Gill, P.; Shan, W.; Balog, A.; Chen, K.; Tebben, A.; Rampulla, R.; et al. Discovery of Clinical Candidate BMS-906024: A Potent-Notch Inhibitor for the Treatment of Leukemia and Solid Tumors. ACS Med. Chem. Lett. 2015, 6, 523. [Google Scholar] [CrossRef] [PubMed]
  335. Ferrarotto, R.; Mishra, V.; Herz, E.; Yaacov, A.; Solomon, O.; Rauch, R.; Mondshine, A.; Motin, M.; Leibovich-Rivkin, T.; Davis, M.; et al. AL101, a Gamma-Secretase Inhibitor, Has Potent Antitumor Activity against Adenoid Cystic Carcinoma with Activated NOTCH Signaling. Cell Death Dis. 2022, 13, 678. [Google Scholar] [CrossRef] [PubMed]
  336. Zweidler-McKay, P.A.; DeAngelo, D.J.; Douer, D.; Dombret, H.; Ottmann, O.G.; Vey, N.; Thomas, D.A.; Zhu, L.; Huang, F.; Bajaj, G.; et al. The Safety and Activity of BMS-906024, a Gamma Secretase Inhibitor (GSI) with Anti-Notch Activity, in Patients with Relapsed T-Cell Acute Lymphoblastic Leukemia (T-ALL): Initial Results of a Phase 1 Trial. Blood 2014, 124, 968. [Google Scholar] [CrossRef]
  337. Ferrarotto, R.; Ho, A.L.; Wirth, L.J.; Dekel, E.; Walker, R.W.; Vergara-Silva, A.L. ACCURACY: Phase (P) 2 Trial of AL101, a Pan-Notch Inhibitor, in Patients (Pts) with Recurrent/Metastatic (R/M) Adenoid Cystic Carcinoma (ACC) with Notch Activating Mutations (Notch Act Mut). J. Clin. Oncol. 2019, 37 (Suppl. S15), TPS6098. [Google Scholar] [CrossRef]
  338. Morgan, K.M.; Fischer, B.S.; Lee, F.Y.; Shah, J.J.; Bertino, J.R.; Rosenfeld, J.; Singh, A.; Khiabanian, H.; Pine, S.R. Gamma Secretase Inhibition by BMS-906024 Enhances Efficacy of Paclitaxel in Lung Adenocarcinoma. Mol. Cancer Ther. 2017, 16, 2759. [Google Scholar] [CrossRef]
  339. Luistro, L.; He, W.; Smith, M.; Packman, K.; Vilenchik, M.; Carvajal, D.; Roberts, J.; Cai, J.; Berkofsky-Fessler, W.; Hilton, H.; et al. Preclinical Profile of a Potent γ-Secretase Inhibitor Targeting Notch Signaling with in Vivo Efficacy and Pharmacodynamic Properties. Cancer Res. 2009, 69, 7672–7680. [Google Scholar] [CrossRef]
  340. Huynh, C.; Poliseno, L.; Segura, M.F.; Medicherla, R.; Haimovic, A.; Menendez, S.; Shang, S.; Pavlick, A.; Shao, Y.; Darvishian, F.; et al. The Novel Gamma Secretase Inhibitor RO4929097 Reduces the Tumor Initiating Potential of Melanoma. PLoS ONE 2011, 6, e25264. [Google Scholar] [CrossRef]
  341. Strosberg, J.R.; Yeatman, T.; Weber, J.; Coppola, D.; Schell, M.J.; Han, G.; Almhanna, K.; Kim, R.; Valone, T.; Jump, H.; et al. A Phase II Study of RO4929097 in Metastatic Colorectal Cancer. Eur. J. Cancer 2012, 48, 997. [Google Scholar] [CrossRef] [PubMed]
  342. Lee, S.M.; Moon, J.; Redman, B.G.; Chidiac, T.; Flaherty, L.E.; Zha, Y.; Othus, M.; Ribas, A.; Sondak, V.K.; Gajewski, T.F.; et al. Phase 2 Study of RO4929097, a Gamma-Secretase Inhibitor, in Metastatic Melanoma: SWOG 0933. Cancer 2015, 121, 432–440. [Google Scholar] [CrossRef] [PubMed]
  343. Gounder, M.M.; Rosenbaum, E.; Wu, N.; Dickson, M.A.; Sheikh, T.N.; D’Angelo, S.P.; Chi, P.; Keohan, M.L.; Erinjeri, J.P.; Antonescu, C.R.; et al. A Phase Ib/II Randomized Study of RO4929097, a Gamma-Secretase or Notch Inhibitor with or without Vismodegib, a Hedgehog Inhibitor, in Advanced Sarcoma. Clin. Cancer Res. 2022, 28, 1586–1594. [Google Scholar] [CrossRef] [PubMed]
  344. De Jesus-Acosta, A.; Laheru, D.; Maitra, A.; Arcaroli, J.; Rudek, M.A.; Dasari, A.; Blatchford, P.J.; Quackenbush, K.; Messersmith, W. A Phase II Study of the Gamma Secretase Inhibitor RO4929097 in Patients with Previously Treated Metastatic Pancreatic Adenocarcinoma. Investig. New Drugs 2014, 32, 739. [Google Scholar] [CrossRef] [PubMed]
  345. Wu, C.X.; Xu, A.; Zhang, C.C.; Olson, P.; Chen, L.; Lee, T.K.; Cheung, T.T.; Lo, C.M.; Wang, X.Q. Notch Inhibitor PF-03084014 Inhibits Hepatocellular Carcinoma Growth and Metastasis via Suppression of Cancer Stemness Due to Reduced Activation of Notch1–Stat3. Mol. Cancer Ther. 2017, 16, 1531–1543. [Google Scholar] [CrossRef] [PubMed]
  346. Cui, D.; Dai, J.; Keller, J.M.; Mizokami, A.; Xia, S.; Keller, E.T. Notch Pathway Inhibition Using PF-03084014, a γ-Secretase Inhibitor (GSI), Enhances the Anti-Tumor Effect of Docetaxel in Prostate Cancer. Clin. Cancer Res. 2015, 21, 4619. [Google Scholar] [CrossRef]
  347. Kummar, S.; Coyne, G.O.S.; Do, K.T.; Turkbey, B.; Meltzer, P.S.; Polley, E.; Choyke, P.L.; Meehan, R.; Vilimas, R.; Horneffer, Y.; et al. Clinical Activity of the γ-Secretase Inhibitor PF-03084014 in Adults with Desmoid Tumors (Aggressive Fibromatosis). J. Clin. Oncol. 2017, 35, 1561. [Google Scholar] [CrossRef] [PubMed]
  348. Gounder, M.; Ratan, R.; Alcindor, T.; Schöffski, P.; van der Graaf, W.T.; Wilky, B.A.; Riedel, R.F.; Lim, A.; Smith, L.M.; Moody, S.; et al. Nirogacestat, a γ-Secretase Inhibitor for Desmoid Tumors. N. Engl. J. Med. 2023, 388, 898–912. [Google Scholar] [CrossRef]
  349. Locatelli, M.A.; Aftimos, P.; Claire Dees, E.; LoRusso, P.M.; Pegram, M.D.; Awada, A.; Huang, B.; Cesari, R.; Jiang, Y.; Shaik, M.N.; et al. Phase I Study of the Gamma Secretase Inhibitor PF-03084014 in Combination with Docetaxel in Patients with Advanced Triple-Negative Breast Cancer. Oncotarget 2017, 8, 2320. [Google Scholar] [CrossRef]
  350. Dai, G.; Deng, S.; Guo, W.; Yu, L.; Yang, J.; Zhou, S.; Gao, T. Notch Pathway Inhibition Using DAPT, a γ-Secretase Inhibitor (GSI), Enhances the Antitumor Effect of Cisplatin in Resistant Osteosarcoma. Mol. Carcinog. 2019, 58, 3–18. [Google Scholar] [CrossRef]
  351. Barat, S.; Chen, X.; Bui, K.C.; Bozko, P.; Götze, J.; Christgen, M.; Krech, T.; Malek, N.P.; Plentz, R.R. Gamma-Secretase Inhibitor IX (GSI) Impairs Concomitant Activation of Notch and Wnt-Beta-Catenin Pathways in CD44+ Gastric Cancer Stem Cells. Stem Cells Transl. Med. 2017, 6, 819. [Google Scholar] [CrossRef]
  352. Feng, J.; Wang, J.; Liu, Q.; Li, J.; Zhang, Q.; Zhuang, Z.; Yao, X.; Liu, C.; Li, Y.; Cao, L.; et al. DAPT, a γ-Secretase Inhibitor, Suppresses Tumorigenesis, and Progression of Growth Hormone-Producing Adenomas by Targeting Notch Signaling. Front. Oncol. 2019, 9, 809. [Google Scholar] [CrossRef]
  353. Schott, A.F.; Landis, M.D.; Dontu, G.; Griffith, K.A.; Layman, R.M.; Krop, I.; Paskett, L.A.; Wong, H.; Dobrolecki, L.E.; Lewis, M.T.; et al. Preclinical and Clinical Studies of Gamma Secretase Inhibitors with Docetaxel on Human Breast Tumors. Clin. Cancer Res. 2013, 19, 1512. [Google Scholar] [CrossRef]
  354. Chen, X.; Gong, L.; Ou, R.; Zheng, Z.; Chen, J.; Xie, F.; Huang, X.; Qiu, J.; Zhang, W.; Jiang, Q.; et al. Sequential Combination Therapy of Ovarian Cancer with Cisplatin and γ-Secretase Inhibitor MK-0752. Gynecol. Oncol. 2016, 140, 537–544. [Google Scholar] [CrossRef]
  355. Krop, I.; Demuth, T.; Guthrie, T.; Wen, P.Y.; Mason, W.P.; Chinnaiyan, P.; Butowski, N.; Groves, M.D.; Kesari, S.; Freedman, S.J.; et al. Phase I Pharmacologic and Pharmacodynamic Study of the Gamma Secretase (Notch) Inhibitor MK-0752 in Adult Patients with Advanced Solid Tumors. J. Clin. Oncol. 2012, 30, 2307–2313. [Google Scholar] [CrossRef]
  356. Cook, N.; Basu, B.; Smith, D.M.; Gopinathan, A.; Evans, J.; Steward, W.P.; Palmer, D.; Propper, D.; Venugopal, B.; Hategan, M.; et al. A Phase I Trial of the γ-Secretase Inhibitor MK-0752 in Combination with Gemcitabine in Patients with Pancreatic Ductal Adenocarcinoma. Br. J. Cancer 2018, 118, 793–801. [Google Scholar] [CrossRef]
  357. Hoffman, L.M.; Fouladi, M.; Olson, J.; Daryani, V.M.; Stewart, C.F.; Wetmore, C.; Kocak, M.; Onar-Thomas, A.; Wagner, L.; Gururangan, S.; et al. Phase I Trial of Weekly MK-0752 in Children with Refractory Central Nervous System Malignancies: A Pediatric Brain Tumor Consortium Study. Childs Nerv. Syst. 2015, 31, 1283. [Google Scholar] [CrossRef]
  358. Habets, R.A.; De Bock, C.E.; Serneels, L.; Lodewijckx, I.; Verbeke, D.; Nittner, D.; Narlawar, R.; Demeyer, S.; Dooley, J.; Liston, A.; et al. Safe Targeting of T Cell Acute Lymphoblastic Leukemia by Pathology-Specific NOTCH Inhibition. Sci. Transl. Med. 2019, 11, 6246. [Google Scholar] [CrossRef]
  359. Lehal, R.; Zaric, J.; Vigolo, M.; Urech, C.; Frismantas, V.; Zangger, N.; Cao, L.; Berger, A.; Chicote, I.; Loubéry, S.; et al. Pharmacological Disruption of the Notch Transcription Factor Complex. Proc. Natl. Acad. Sci. USA 2020, 117, 16292–16301. [Google Scholar] [CrossRef]
  360. Vigolo, M.; Urech, C.; Lamy, S.; Monticone, G.; Zabaleta, J.; Hossain, F.; Wyczechowska, D.; Del Valle, L.; O’Regan, R.M.; Miele, L.; et al. The Efficacy of CB-103, a First-in-Class Transcriptional Notch Inhibitor, in Preclinical Models of Breast Cancer. Cancers 2023, 15, 3957. [Google Scholar] [CrossRef] [PubMed]
  361. Lu, H.Y.; Zu, Y.X.; Jiang, X.W.; Sun, X.T.; Liu, T.Y.; Li, R.L.; Wu, Q.; Zhang, Y.S.; Zhao, Q.C. Novel ADAM-17 Inhibitor ZLDI-8 Inhibits the Proliferation and Metastasis of Chemo-Resistant Non-Small-Cell Lung Cancer by Reversing Notch and Epithelial Mesenchymal Transition in Vitro and in Vivo. Pharmacol. Res. 2019, 148, 104406. [Google Scholar] [CrossRef]
  362. Zhang, Y.; Li, D.; Jiang, Q.; Cao, S.; Sun, H.; Chai, Y.; Li, X.; Ren, T.; Yang, R.; Feng, F.; et al. Novel ADAM-17 Inhibitor ZLDI-8 Enhances the in Vitro and in Vivo Chemotherapeutic Effects of Sorafenib on Hepatocellular Carcinoma Cells. Cell Death Dis. 2018, 9, 743. [Google Scholar] [CrossRef] [PubMed]
  363. Li, D.D.; Zhao, C.H.; Ding, H.W.; Wu, Q.; Ren, T.S.; Wang, J.; Chen, C.Q.; Zhao, Q.C. A Novel Inhibitor of ADAM17 Sensitizes Colorectal Cancer Cells to 5-Fluorouracil by Reversing Notch and Epithelial-Mesenchymal Transition in Vitro and in Vivo. Cell Prolif. 2018, 51, e12480. [Google Scholar] [CrossRef] [PubMed]
  364. Sharma, A.; Paranjape, A.N.; Rangarajan, A.; Dighe, R.R. A Monoclonal Antibody against Human Notch1 Ligand-Binding Domain Depletes Subpopulation of Putative Breast Cancer Stem-like Cells. Mol. Cancer Ther. 2012, 11, 77–86. [Google Scholar] [CrossRef]
  365. Wu, Y.; Cain-Hom, C.; Choy, L.; Hagenbeek, T.J.; De Leon, G.P.; Chen, Y.; Finkle, D.; Venook, R.; Wu, X.; Ridgway, J.; et al. Therapeutic Antibody Targeting of Individual Notch Receptors. Nature 2010, 464, 1052–1057. [Google Scholar] [CrossRef]
  366. Yen, W.C.; Fischer, M.M.; Axelrod, F.; Bond, C.; Cain, J.; Cancilla, B.; Henner, W.R.; Meisner, R.; Sato, A.; Shah, J.; et al. Targeting Notch Signaling with a Notch2/Notch3 Antagonist (Tarextumab) Inhibits Tumor Growth and Decreases Tumor-Initiating Cell Frequency. Clin. Cancer Res. 2015, 21, 2084–2095. [Google Scholar] [CrossRef]
  367. Pietanza, M.C.; Spira, A.I.; Jotte, R.M.; Gadgeel, S.M.; Mita, A.C.; Hart, L.L.; Gluck, W.L.; Chiang, A.C.; Liu, S.V.; Kapoun, A.M.; et al. Final Results of Phase Ib of Tarextumab (TRXT, OMP-59R5, Anti-Notch2/3) in Combination with Etoposide and Platinum (EP) in Patients (Pts) with Untreated Extensive-Stage Small-Cell Lung Cancer (ED-SCLC). J. Clin. Oncol. 2015, 33 (Suppl. S15), 7508. [Google Scholar] [CrossRef]
  368. Smith, D.C.; Chugh, R.; Patnaik, A.; Papadopoulos, K.P.; Wang, M.; Kapoun, A.M.; Xu, L.; Dupont, J.; Stagg, R.J.; Tolcher, A. A Phase 1 Dose Escalation and Expansion Study of Tarextumab (OMP-59R5) in Patients with Solid Tumors. Investig. New Drugs 2019, 37, 722. [Google Scholar] [CrossRef]
  369. Hu, Z.I.; Bendell, J.C.; Bullock, A.; LoConte, N.K.; Hatoum, H.; Ritch, P.; Hool, H.; Leach, J.W.; Sanchez, J.; Sohal, D.P.S.; et al. A Randomized Phase II Trial of Nab-Paclitaxel and Gemcitabine with Tarextumab or Placebo in Patients with Untreated Metastatic Pancreatic Cancer. Cancer Med. 2019, 8, 5148–5157. [Google Scholar] [CrossRef]
  370. Ferrarotto, R.; Eckhardt, G.; Patnaik, A.; LoRusso, P.; Faoro, L.; Heymach, J.V.; Kapoun, A.M.; Xu, L.; Munster, P. A Phase I Dose-Escalation and Dose-Expansion Study of Brontictuzumab in Subjects with Selected Solid Tumors. Ann. Oncol. 2018, 29, 1561–1568. [Google Scholar] [CrossRef] [PubMed]
  371. Casulo, C.; Ruan, J.; Dang, N.H.; Gore, L.; Diefenbach, C.; Beaven, A.W.; Castro, J.E.; Porcu, P.; Faoro, L.; Dupont, J.; et al. Safety and Preliminary Efficacy Results of a Phase I First-in-Human Study of the Novel Notch-1 Targeting Antibody Brontictuzumab (OMP-52M51) Administered Intravenously to Patients with Hematologic Malignancies. Blood 2016, 128, 5108. [Google Scholar] [CrossRef]
  372. Rosen, L.S.; Wesolowski, R.; Baffa, R.; Liao, K.H.; Hua, S.Y.; Gibson, B.L.; Pirie-Shepherd, S.; Tolcher, A.W. A Phase I, Dose-Escalation Study of PF-06650808, an Anti-Notch3 Antibody–Drug Conjugate, in Patients with Breast Cancer and Other Advanced Solid Tumors. Investig. New Drugs 2020, 38, 120–130. [Google Scholar] [CrossRef] [PubMed]
  373. Chiorean, E.G.; LoRusso, P.; Strother, R.M.; Diamond, J.R.; Younger, A.; Messersmith, W.A.; Adriaens, L.; Liu, L.; Kao, R.J.; DiCioccio, A.T.; et al. A Phase I First-in-Human Study of Enoticumab (REGN421), a Fully Human Delta-like Ligand 4 (Dll4) Monoclonal Antibody in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2015, 21, 2695–2703. [Google Scholar] [CrossRef] [PubMed]
  374. Coleman, R.L.; Handley, K.F.; Burger, R.; Molin, G.Z.D.; Stagg, R.; Sood, A.K.; Moore, K.N. Demcizumab Combined with Paclitaxel for Platinum-Resistant Ovarian, Primary Peritoneal, and Fallopian Tube Cancer: The SIERRA Open-Label Phase Ib Trial. Gynecol. Oncol. 2020, 157, 386–391. [Google Scholar] [CrossRef] [PubMed]
  375. McKeage, M.J.; Kotasek, D.; Markman, B.; Hidalgo, M.; Millward, M.J.; Jameson, M.B.; Harris, D.L.; Stagg, R.J.; Kapoun, A.M.; Xu, L.; et al. Phase IB Trial of the Anti-Cancer Stem Cell DLL4-Binding Agent Demcizumab with Pemetrexed and Carboplatin as First-Line Treatment of Metastatic Non-Squamous NSCLC. Target. Oncol. 2018, 13, 89–98. [Google Scholar] [CrossRef] [PubMed]
  376. Jenkins, D.W.; Ross, S.; Veldman-Jones, M.; Foltz, I.N.; Clavette, B.C.; Manchulenko, K.; Eberlein, C.; Kendrew, J.; Petteruti, P.; Cho, S.; et al. MEDI0639: A Novel Therapeutic Antibody Targeting Dll4 Modulates Endothelial Cell Function and Angiogenesis in Vivo. Mol. Cancer Ther. 2012, 11, 1650–1660. [Google Scholar] [CrossRef] [PubMed]
  377. Falchook, G.S.; Dowlati, A.; Naing, A.; Gribbin, M.J.; Jenkins, D.W.; Chang, L.L.; Lai, D.W.; Smith, D.C. Phase I Study of MEDI0639 in Patients with Advanced Solid Tumors. J. Clin. Oncol. 2015, 33 (Suppl. S15), 3024. [Google Scholar] [CrossRef]
  378. Xu, Z.; Wang, Z.; Jia, X.; Wang, L.; Chen, Z.; Wang, S.; Wang, M.; Zhang, J.; Wu, M. MMGZ01, an Anti-DLL4 Monoclonal Antibody, Promotes Nonfunctional Vessels and Inhibits Breast Tumor Growth. Cancer Lett. 2016, 372, 118–127. [Google Scholar] [CrossRef]
  379. Jimeno, A.; Moore, K.N.; Gordon, M.; Chugh, R.; Diamond, J.R.; Aljumaily, R.; Mendelson, D.; Kapoun, A.M.; Xu, L.; Stagg, R.; et al. A First-in-Human Phase 1a Study of the Bispecific Anti-DLL4/Anti-VEGF Antibody Navicixizumab (OMP-305B83) in Patients with Previously Treated Solid Tumors. Investig. New Drugs 2019, 37, 461–472. [Google Scholar] [CrossRef]
  380. Fu, S.; Corr, B.R.; Culm-Merdek, K.; Mockbee, C.; Youssoufian, H.; Stagg, R.; Naumann, R.W.; Wenham, R.M.; Rosengarten, R.D.; Benjamin, L.; et al. Phase Ib Study of Navicixizumab Plus Paclitaxel in Patients with Platinum-Resistant Ovarian, Primary Peritoneal, or Fallopian Tube Cancer. J. Clin. Oncol. 2022, 40, 2568. [Google Scholar] [CrossRef] [PubMed]
  381. Li, Y.; Hickson, J.A.; Ambrosi, D.J.; Haasch, D.L.; Foster-Duke, K.D.; Eaton, L.J.; DiGiammarino, E.L.; Panchal, S.C.; Jiang, F.; Mudd, S.R.; et al. Abt-165, a Dual Variable Domain Immunoglobulin (Dvd-Ig) Targeting Dll4 and Vegf, Demonstrates Superior Efficacy and Favorable Safety Profiles in Preclinical Models. Mol. Cancer Ther. 2018, 17, 1039–1050. [Google Scholar] [CrossRef] [PubMed]
  382. Wainberg, Z.; Strickler, J.; Gordon, M.; Barve, M.; Wang, L.; Yue, H.; Motwani, M.; Kasichayanula, S.; Naumovski, L.; Hamilton, E. Phase 1b Open-Label Study Evaluating the Safety, Pharmacokinetics, and Preliminary Efficacy of ABT-165 plus FOLFIRI in Patients with Second-Line (2L) Colorectal Cancer (CRC). Ann. Oncol. 2018, 29, v66. [Google Scholar] [CrossRef]
  383. Giffin, M.J.; Cooke, K.; Lobenhofer, E.K.; Estrada, J.; Zhan, J.; Deegen, P.; Thomas, M.; Murawsky, C.M.; Werner, J.; Liu, S.; et al. AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer. Clin. Cancer Res. 2021, 27, 1526–1537. [Google Scholar] [CrossRef] [PubMed]
  384. Paz-Ares, L.; Champiat, S.; Lai, W.V.; Izumi, H.; Govindan, R.; Boyer, M.; Hummel, H.D.; Borghaei, H.; Johnson, M.L.; Steeghs, N.; et al. Tarlatamab, a First-in-Class DLL3-Targeted Bispecific T-Cell Engager, in Recurrent Small-Cell Lung Cancer: An Open-Label, Phase I Study. J. Clin. Oncol. 2023, 41, 2893. [Google Scholar] [CrossRef] [PubMed]
  385. Ahn, M.-J.; Cho, B.C.; Felip, E.; Korantzis, I.; Ohashi, K.; Majem, M.; Juan-Vidal, O.; Handzhiev, S.; Izumi, H.; Lee, J.-S.; et al. Tarlatamab for Patients with Previously Treated Small-Cell Lung Cancer. N. Engl. J. Med. 2023, 389, 2063–2075. [Google Scholar] [CrossRef]
  386. Johnson, M.L.; Dy, G.K.; Mamdani, H.; Dowlati, A.; Schoenfeld, A.J.; Pacheco, J.M.; Sanborn, R.E.; Menon, S.P.; Santiago, L.; Yaron, Y.; et al. Interim Results of an Ongoing Phase 1/2a Study of HPN328, a Tri-Specific, Half-Life Extended, DLL3-Targeting, T-Cell Engager, in Patients with Small Cell Lung Cancer and Other Neuroendocrine Cancers. J. Clin. Oncol. 2022, 40 (Suppl. S16), 8566. [Google Scholar] [CrossRef]
  387. Hann, C.L.; Burns, T.F.; Dowlati, A.; Morgensztern, D.; Ward, P.J.; Koch, M.M.; Chen, C.; Ludwig, C.; Patel, M.; Nimeiri, H.; et al. A Phase 1 Study Evaluating Rovalpituzumab Tesirine in Frontline Treatment of Patients with Extensive-Stage SCLC. J. Thorac. Oncol. 2021, 16, 1582–1588. [Google Scholar] [CrossRef] [PubMed]
  388. Saunders, L.R.; Bankovich, A.J.; Anderson, W.C.; Aujay, M.A.; Bheddah, S.; Black, K.A.; Desai, R.; Escarpe, P.A.; Hampl, J.; Laysang, A.; et al. A DLL3-Targeted Antibody-Drug Conjugate Eradicates High-Grade Pulmonary Neuroendocrine Tumor-Initiating Cells in Vivo. Sci. Transl. Med. 2015, 7, 302ra136. [Google Scholar] [CrossRef]
  389. Johnson, M.L.; Zvirbule, Z.; Laktionov, K.; Helland, A.; Cho, B.C.; Gutierrez, V.; Colinet, B.; Lena, H.; Wolf, M.; Gottfried, M.; et al. Rovalpituzumab Tesirine as a Maintenance Therapy After First-Line Platinum-Based Chemotherapy in Patients with Extensive-Stage–SCLC: Results From the Phase 3 MERU Study. J. Thorac. Oncol. 2021, 16, 1570–1581. [Google Scholar] [CrossRef]
  390. Morgensztern, D.; Besse, B.; Greillier, L.; Santana-Davila, R.; Ready, N.; Hann, C.L.; Glisson, B.S.; Farago, A.F.; Dowlati, A.; Rudin, C.M.; et al. Efficacy and Safety of Rovalpituzumab Tesirine in Third-Line and beyond Patients with DLL3-Expressing, Relapsed/Refractory Small-Cell Lung Cancer: Results from the Phase II TrINITY Study. Clin. Cancer Res. 2019, 25, 6958–6966. [Google Scholar] [CrossRef] [PubMed]
  391. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/β-Catenin Signaling: Function, Biological Mechanisms, and Therapeutic Opportunities. Signal Transduct. Target. Ther. 2022, 7, 3. [Google Scholar] [CrossRef]
  392. Clevers, H. Wnt/β-Catenin Signaling in Development and Disease. Cell 2006, 127, 469–480. [Google Scholar] [CrossRef] [PubMed]
  393. Yu, F.; Yu, C.; Li, F.; Zuo, Y.; Wang, Y.; Yao, L.; Wu, C.; Wang, C.; Ye, L. Wnt/β-Catenin Signaling in Cancers and Targeted Therapies. Signal Transduct. Target. Ther. 2021, 6, 307. [Google Scholar] [CrossRef] [PubMed]
  394. Willert, K.; Brown, J.D.; Danenberg, E.; Duncan, A.W.; Weissman, I.L.; Reya, T.; Yates, J.R.; Nusse, R. Wnt Proteins Are Lipid-Modified and Can Act as Stem Cell Growth Factors. Nature 2003, 423, 448–452. [Google Scholar] [CrossRef] [PubMed]
  395. Baarsma, H.A.; Königshoff, M. ‘WNT-Er Is Coming’: WNT Signaling in Chronic Lung Diseases. Thorax 2017, 72, 746–759. [Google Scholar] [CrossRef] [PubMed]
  396. Liu, Y.; Wang, W.-M.; Zhang, X.-L.; He, H.-Q.; Sun, X.-L.; Zeng, H.; Xu, X.-F.; Huang, L.; Zhu, Z.; Zhang, L.; et al. AGE/RAGE Promotes Thecalcification of Human Aortic Smooth Muscle Cells via the Wnt/β-Catenin Axis. Am. J. Transl. Res. 2016, 8, 4644. [Google Scholar] [PubMed]
  397. González-Fernández, C.; Gonzalez, P.; Andres-Benito, P.; Ferrer, I.; Rodríguez, F.J. Wnt Signaling Alterations in the Human Spinal Cord of Amyotrophic Lateral Sclerosis Cases: Spotlight on Fz2 and Wnt5a. Mol. Neurobiol. 2019, 56, 6777–6791. [Google Scholar] [CrossRef]
  398. Vermeulen, L.; De Sousa E Melo, F.; Van Der Heijden, M.; Cameron, K.; De Jong, J.H.; Borovski, T.; Tuynman, J.B.; Todaro, M.; Merz, C.; Rodermond, H.; et al. Wnt Activity Defines Colon Cancer Stem Cells and Is Regulated by the Microenvironment. Nat. Cell Biol. 2010, 12, 468–476. [Google Scholar] [CrossRef]
  399. Guo, W.; Lasky, J.L.; Chang, C.J.; Mosessian, S.; Lewis, X.; Xiao, Y.; Yeh, J.E.; Chen, J.Y.; Iruela-Arispe, M.L.; Varella-Garcia, M.; et al. Multi-Genetic Events Collaboratively Contribute to Pten-Null Leukaemia Stem-Cell Formation. Nature 2008, 453, 529–533. [Google Scholar] [CrossRef]
  400. Jang, G.B.; Kim, J.Y.; Cho, S.D.; Park, K.S.; Jung, J.Y.; Lee, H.Y.; Hong, I.S.; Nam, J.S. Blockade of Wnt/β-Catenin Signaling Suppresses Breast Cancer Metastasis by Inhibiting CSC-like Phenotype. Sci. Rep. 2015, 5, 12465. [Google Scholar] [CrossRef] [PubMed]
  401. Lee, S.H.; Koo, B.S.; Kim, J.M.; Huang, S.; Rho, Y.S.; Bae, W.J.; Kang, H.J.; Kim, Y.S.; Moon, J.H.; Lim, Y.C. Wnt/β-Catenin Signaling Maintains Self-Renewal and Tumorigenicity of Head and Neck Squamous Cell Carcinoma Stem-like Cells by Activating Oct4. J. Pathol. 2014, 234, 99–107. [Google Scholar] [CrossRef]
  402. Gurney, A.; Axelrod, F.; Bond, C.J.; Cain, J.; Chartier, C.; Donigan, L.; Fischer, M.; Chaudhari, A.; Ji, M.; Kapoun, A.M.; et al. Wnt Pathway Inhibition via the Targeting of Frizzled Receptors Results in Decreased Growth and Tumorigenicity of Human Tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 11717–11722. [Google Scholar] [CrossRef] [PubMed]
  403. Smith, D.C.; Rosen, L.S.; Chugh, R.; Goldman, J.W.; Xu, L.; Kapoun, A.; Brachmann, R.K.; Dupont, J.; Stagg, R.J.; Tolcher, A.W.; et al. First-in-Human Evaluation of the Human Monoclonal Antibody Vantictumab (OMP-18R5; Anti-Frizzled) Targeting the WNT Pathway in a Phase I Study for Patients with Advanced Solid Tumors. J. Clin. Oncol. 2013, 31 (Suppl. S15), 2540. [Google Scholar] [CrossRef]
  404. Davis, S.L.; Cardin, D.B.; Shahda, S.; Lenz, H.-J.; Dotan, E.; O’Neil, B.; Kapoun, A.M.; Stagg, R.J.; Berlin, J.; Messersmith, W.A.; et al. A Phase Ib Dose Escalation Study of Vantictumab (VAN) in Combination with Nab-Paclitaxel (Nab-P) and Gemcitabine (G) in Patients with Previously Untreated Stage IV Pancreatic Cancer. J. Clin. Oncol. 2019, 37 (Suppl. S4), 249. [Google Scholar] [CrossRef]
  405. Diamond, J.R.; Becerra, C.; Richards, D.; Mita, A.; Osborne, C.; O’Shaughnessy, J.; Zhang, C.; Henner, R.; Kapoun, A.M.; Xu, L.; et al. Phase Ib Clinical Trial of the Anti-Frizzled Antibody Vantictumab (OMP-18R5) plus Paclitaxel in Patients with Locally Advanced or Metastatic HER2-Negative Breast Cancer. Breast Cancer Res. Treat. 2020, 184, 53–62. [Google Scholar] [CrossRef] [PubMed]
  406. Pavlovic, Z.; Adams, J.J.; Blazer, L.L.; Gakhal, A.K.; Jarvik, N.; Steinhart, Z.; Robitaille, M.; Mascall, K.; Pan, J.; Angers, S.; et al. A Synthetic Anti-Frizzled Antibody Engineered for Broadened Specificity Exhibits Enhanced Anti-Tumor Properties. MAbs 2018, 10, 1157–1167. [Google Scholar] [CrossRef] [PubMed]
  407. Steinhart, Z.; Pavlovic, Z.; Chandrashekhar, M.; Hart, T.; Wang, X.; Zhang, X.; Robitaille, M.; Brown, K.R.; Jaksani, S.; Overmeer, R.; et al. Genome-Wide CRISPR Screens Reveal a Wnt–FZD5 Signaling Circuit as a Druggable Vulnerability of RNF43-Mutant Pancreatic Tumors. Nat. Med. 2016, 23, 60–68. [Google Scholar] [CrossRef] [PubMed]
  408. Li, H.K.; Sugyo, A.; Tsuji, A.B.; Morokoshi, Y.; Minegishi, K.; Nagatsu, K.; Kanda, H.; Harada, Y.; Nagayama, S.; Katagiri, T.; et al. α-Particle Therapy for Synovial Sarcoma in the Mouse Using an Astatine-211-Labeled Antibody against Frizzled Homolog 10. Cancer Sci. 2018, 109, 2302–2309. [Google Scholar] [CrossRef]
  409. Sudo, H.; Tsuji, A.B.; Sugyo, A.; Harada, Y.; Nagayama, S.; Katagiri, T.; Nakamura, Y.; Higashi, T. FZD10-targeted A-radioimmunotherapy with 225Ac-labeled OTSA101 Achieves Complete Remission in a Synovial Sarcoma Model. Cancer Sci. 2022, 113, 721. [Google Scholar] [CrossRef]
  410. Giraudet, A.-L.; Badel, J.-N.; Cassier, P.; Desuzinges, C.; Kriza, D.; Perol, D.; Blay, J.-Y. SYNFRIZZ-A Phase Ia/Ib of a Radiolabelled Monoclonal AB for the Treatment of Relapsing Synovial Sarcoma. J. Nucl. Med. 2014, 55 (Suppl. S1), 223. [Google Scholar]
  411. Giraudet, A.L.; Cassier, P.A.; Iwao-Fukukawa, C.; Garin, G.; Badel, J.N.; Kryza, D.; Chabaud, S.; Gilles-Afchain, L.; Clapisson, G.; Desuzinges, C.; et al. A First-in-Human Study Investigating Biodistribution, Safety and Recommended Dose of a New Radiolabeled MAb Targeting FZD10 in Metastatic Synovial Sarcoma Patients. BMC Cancer 2018, 18, 646. [Google Scholar] [CrossRef]
  412. Le, P.N.; McDermott, J.D.; Jimeno, A. Targeting the Wnt Pathway in Human Cancers: Therapeutic Targeting with a Focus on OMP-54F28. Pharmacol. Ther. 2015, 146, 1–11. [Google Scholar] [CrossRef]
  413. Fischer, M.M.; Cancilla, B.; Yeung, V.P.; Cattaruzza, F.; Chartier, C.; Murriel, C.L.; Cain, J.; Tam, R.; Cheng, C.Y.; Evans, J.W.; et al. WNT Antagonists Exhibit Unique Combinatorial Antitumor Activity with Taxanes by Potentiating Mitotic Cell Death. Sci. Adv. 2017, 3, e1700090. [Google Scholar] [CrossRef]
  414. Jimeno, A.; Gordon, M.; Chugh, R.; Messersmith, W.; Mendelson, D.; Dupont, J.; Stagg, R.; Kapoun, A.M.; Xu, L.; Uttamsingh, S.; et al. A First-in-Human Phase I Study of the Anticancer Stem Cell Agent Ipafricept (OMP-54F28), a Decoy Receptor for Wnt Ligands, in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2017, 23, 7490–7497. [Google Scholar] [CrossRef]
  415. Moore, K.N.; Gunderson, C.C.; Sabbatini, P.; McMeekin, D.S.; Mantia-Smaldone, G.; Burger, R.A.; Morgan, M.A.; Kapoun, A.M.; Brachmann, R.K.; Stagg, R.; et al. A Phase 1b Dose Escalation Study of Ipafricept (OMP54F28) in Combination with Paclitaxel and Carboplatin in Patients with Recurrent Platinum-Sensitive Ovarian Cancer. Gynecol. Oncol. 2019, 154, 294–301. [Google Scholar] [CrossRef]
  416. Dotan, E.; Cardin, D.B.; Lenz, H.J.; Messersmith, W.; O’Neil, B.; Cohen, S.J.; Denlinger, C.S.; Shahda, S.; Astsaturov, I.; Kapoun, A.M.; et al. Phase Ib Study of Wnt Inhibitor Ipafricept with Gemcitabine and Nab-Paclitaxel in Patients with Previously Untreated Stage IV Pancreatic Cancer. Clin. Cancer Res. 2020, 26, 5348–5357. [Google Scholar] [CrossRef]
  417. He, B.; You, L.; Uematsu, K.; Xu, Z.; Lee, A.Y.; Matsangou, M.; McCormick, F.; Jablons, D.M. A Monoclonal Antibody against Wnt-1 Induces Apoptosis in Human Cancer Cells. Neoplasia 2004, 6, 7. [Google Scholar] [CrossRef]
  418. Salik, B.; Yi, H.; Hassan, N.; Santiappillai, N.; Vick, B.; Connerty, P.; Duly, A.; Trahair, T.; Woo, A.J.; Beck, D.; et al. Targeting RSPO3-LGR4 Signaling for Leukemia Stem Cell Eradication in Acute Myeloid Leukemia. Cancer Cell 2020, 38, 263–278.e6. [Google Scholar] [CrossRef]
  419. Bendell, J.; Eckhardt, G.S.; Hochster, H.S.; Morris, V.K.; Strickler, J.; Kapoun, A.M.; Wang, M.; Xu, L.; McGuire, K.; Dupont, J.; et al. Initial Results from a Phase 1a/b Study of OMP-131R10, a First-in-Class Anti-RSPO3 Antibody, in Advanced Solid Tumors and Previously Treated Metastatic Colorectal Cancer (CRC). Eur. J. Cancer 2016, 69, S29–S30. [Google Scholar] [CrossRef]
  420. Liu, J.; Pan, S.; Hsieh, M.H.; Ng, N.; Sun, F.; Wang, T.; Kasibhatla, S.; Schuller, A.G.; Li, A.G.; Cheng, D.; et al. Targeting Wnt-Driven Cancer through the Inhibition of Porcupine by LGK974. Proc. Natl. Acad. Sci. USA 2013, 110, 20224–20229. [Google Scholar] [CrossRef] [PubMed]
  421. Han, T.; Schatoff, E.M.; Murphy, C.; Zafra, M.P.; Wilkinson, J.E.; Elemento, O.; Dow, L.E. R-Spondin Chromosome Rearrangements Drive Wnt-Dependent Tumor Initiation and Maintenance in the Intestine. Nat. Commun. 2017, 8, 15945. [Google Scholar] [CrossRef] [PubMed]
  422. Bagheri, M.; Tabatabae Far, M.A.; Mirzaei, H.; Ghasemi, F. Evaluation of Antitumor Effects of Aspirin and LGK974 Drugs on Cellular Signaling Pathways, Cell Cycle and Apoptosis in Colorectal Cancer Cell Lines Compared to Oxaliplatin Drug. Fundam. Clin. Pharmacol. 2020, 34, 51–64. [Google Scholar] [CrossRef] [PubMed]
  423. Rodon, J.; Argilés, G.; Connolly, R.M.; Vaishampayan, U.; de Jonge, M.; Garralda, E.; Giannakis, M.; Smith, D.C.; Dobson, J.R.; McLaughlin, M.E.; et al. Phase 1 Study of Single-Agent WNT974, a First-in-Class Porcupine Inhibitor, in Patients with Advanced Solid Tumors. Br. J. Cancer 2021, 125, 28–37. [Google Scholar] [CrossRef]
  424. Tabernero, J.; Van Cutsem, E.; Garralda, E.; Tai, D.; De Braud, F.; Geva, R.; van Bussel, M.T.J.; Fiorella Dotti, K.; Elez, E.; de Miguel, M.J.; et al. A Phase Ib/II Study of WNT974 + Encorafenib + Cetuximab in Patients with BRAF V600E-Mutant KRAS Wild-Type Metastatic Colorectal Cancer. Oncologist 2023, 28, 230–238. [Google Scholar] [CrossRef] [PubMed]
  425. Madan, B.; Ke, Z.; Harmston, N.; Ho, S.Y.; Frois, A.O.; Alam, J.; Jeyaraj, D.A.; Pendharkar, V.; Ghosh, K.; Virshup, I.H.; et al. Wnt Addiction of Genetically Defined Cancers Reversed by PORCN Inhibition. Oncogene 2016, 35, 2197–2207. [Google Scholar] [CrossRef] [PubMed]
  426. Ng, M.; Tan, D.S.; Subbiah, V.; Weekes, C.D.; Teneggi, V.; Diermayr, V.; Ethirajulu, K.; Yeo, P.; Chen, D.; Blanchard, S.; et al. First-in-Human Phase 1 Study of ETC-159 an Oral PORCN Inhbitor in Patients with Advanced Solid Tumors. J. Clin. Oncol. 2017, 35 (Suppl. S15), 2584. [Google Scholar] [CrossRef]
  427. Tan, D.S.P.; Ng, M.C.H.; Subbiah, V.; Messersmith, W.A.; Strickler, J.H.; Diermayr, V.; Cometa, J.; Blanchard, S.; Nellore, R.; Pendharkar, V.; et al. A Phase 1B Dose Escalation Study of ETC-159 in Combination with Pembrolizumab in Advanced or Metastatic Solid Tumors. J. Clin. Oncol. 2023, 41 (Suppl. S16), 2601. [Google Scholar] [CrossRef]
  428. Proffitt, K.D.; Madan, B.; Ke, Z.; Pendharkar, V.; Ding, L.; Lee, M.A.; Hannoush, R.N.; Virshup, D.M. Pharmacological Inhibition of the Wnt Acyltransferase PORCN Prevents Growth of WNT-Driven Mammary Cancer. Cancer Res. 2013, 73, 502–507. [Google Scholar] [CrossRef]
  429. Cheng, Y.; Phoon, Y.P.; Jin, X.; Chong, S.Y.S.; Ip, J.C.Y.; Wong, B.W.Y.; Lung, M.L. Wnt-C59 Arrests Stemness and Suppresses Growth of Nasopharyngeal Carcinoma in Mice by Inhibiting the Wnt Pathway in the Tumor Microenvironment. Oncotarget 2015, 6, 14428. [Google Scholar] [CrossRef]
  430. Phillips, C.; Bhamra, I.; Eagle, C.; Flanagan, E.; Armer, R.; Jones, C.D.; Bingham, M.; Calcraft, P.; Edmenson Cook, A.; Thompson, B.; et al. The Wnt Pathway Inhibitor RXC004 Blocks Tumor Growth and Reverses Immune Evasion in Wnt Ligand–Dependent Cancer Models. Cancer Res. Commun. 2022, 2, 914–928. [Google Scholar] [CrossRef] [PubMed]
  431. Kelly, J.; Woodcock, S.; Bhamra, I.; Jones, C.; Armer, R.; Robertson, J.; Phillips, C. Abstract 2566: Pre-Clinical Efficacy of the Wnt Pathway Inhibitor RXC004 in Combination with Anti-Cancer Therapies. Cancer Res. 2022, 82 (Suppl. S12), 2566. [Google Scholar] [CrossRef]
  432. Cook, N.; Blagden, S.; Lopez, J.; Sarker, D.; Greystoke, A.; Harris, N.; Kazmi, F.; Naderi, A.; Nintos, G.; Franco, A.O.; et al. 517MO Phase I Study of the Porcupine (PORCN) Inhibitor RXC004 in Patients with Advanced Solid Tumors. Ann. Oncol. 2021, 32, S586–S587. [Google Scholar] [CrossRef]
  433. Giannakis, M.; Le, D.T.; Pishvaian, M.J.; Weinberg, B.A.; Papadopoulos, K.P.; Shen, L.; Gong, J.; Li, J.; Strickler, J.H.; Zhou, A.; et al. Phase 1 Study of WNT Pathway Porcupine Inhibitor CGX1321 and Phase 1b Study of CGX1321 + Pembrolizumab (Pembro) in Patients (Pts) with Advanced Gastrointestinal (GI) Tumors. J. Clin. Oncol. 2023, 41 (Suppl. S16), 3514. [Google Scholar] [CrossRef]
  434. Fiskus, W.; Sharma, S.; Saha, S.; Shah, B.; Devaraj, S.G.T.; Sun, B.; Horrigan, S.; Leveque, C.; Zu, Y.; Iyer, S.; et al. Pre-Clinical Efficacy of Combined Therapy with Novel β-Catenin Antagonist BC2059 and Histone Deacetylase Inhibitor against AML Cells. Leukemia 2015, 29, 1267–1278. [Google Scholar] [CrossRef] [PubMed]
  435. Braggio, D.A.; de Faria, F.C.C.; Koller, D.; Jin, F.; Zewdu, A.; Lopez, G.; Batte, K.; Casadei, L.; Welliver, M.; Horrigan, S.K.; et al. Preclinical Efficacy of the Wnt/β-Catenin Pathway Inhibitor BC2059 for the Treatment of Desmoid Tumors. PLoS ONE 2022, 17, e0276047. [Google Scholar] [CrossRef] [PubMed]
  436. Savvidou, I.; Khong, T.; Cuddihy, A.; McLean, C.; Horrigan, S.; Spencer, A. β-Catenin Inhibitor BC2059 Is Efficacious as Monotherapy or in Combination with Proteasome Inhibitor Bortezomib in Multiple Myeloma. Mol. Cancer Ther. 2017, 16, 1765–1778. [Google Scholar] [CrossRef] [PubMed]
  437. Cranmer, L.D.; Razak, A.R.A.; Ratan, R.; Choy, E.; George, S.; Liebner, D.A.; Stenehjem, D.D.; Gounder, M.M. Results of a Phase I Dose Escalation and Expansion Study of Tegavivint (BC2059), a First-in-Class TBL1 Inhibitor for Patients with Progressive, Unresectable Desmoid Tumor. J. Clin. Oncol. 2022, 40 (Suppl. S16), 11523. [Google Scholar] [CrossRef]
  438. Handeli, S.; Simon, J.A. A Small-Molecule Inhibitor of Tcf/β-Catenin Signaling down-Regulates PPARγ and PPARδ Activities. Mol. Cancer Ther. 2008, 7, 521–529. [Google Scholar] [CrossRef]
  439. Iida, J.; Dorchak, J.; Lehman, J.R.; Clancy, R.; Luo, C.; Chen, Y.; Somiari, S.; Ellsworth, R.E.; Hu, H.; Mural, R.J.; et al. FH535 Inhibited Migration and Growth of Breast Cancer Cells. PLoS ONE 2012, 7, e44418. [Google Scholar] [CrossRef]
  440. Wu, M.Y.; Liang, R.R.; Chen, K.; Shen, M.; Tian, Y.L.; Li, D.M.; Duan, W.M.; Gui, Q.; Gong, F.R.; Lian, L.; et al. FH535 Inhibited Metastasis and Growth of Pancreatic Cancer Cells. Onco Targets Ther. 2015, 8, 1651–1670. [Google Scholar] [CrossRef] [PubMed]
  441. Liu, L.; Zhi, Q.; Shen, M.; Gong, F.R.; Zhou, B.P.; Lian, L.; Shen, B.; Chen, K.; Duan, W.; Wu, M.Y.; et al. FH535, a β-Catenin Pathway Inhibitor, Represses Pancreatic Cancer Xenograft Growth and Angiogenesis. Oncotarget 2016, 7, 47145. [Google Scholar] [CrossRef]
  442. Chen, Y.; Rao, X.; Huang, K.; Jiang, X.; Wang, H.; Teng, L. FH535 Inhibits Proliferation and Motility of Colon Cancer Cells by Targeting Wnt/β-Catenin Signaling Pathway. J. Cancer 2017, 8, 3142. [Google Scholar] [CrossRef] [PubMed]
  443. Perry, J.M.; Tao, F.; Roy, A.; Lin, T.; He, X.C.; Chen, S.; Lu, X.; Nemechek, J.; Ruan, L.; Yu, X.; et al. Overcoming Wnt–β-Catenin Dependent Anticancer Therapy Resistance in Leukaemia Stem Cells. Nat. Cell Biol. 2020, 22, 689–700. [Google Scholar] [CrossRef] [PubMed]
  444. Zhao, Y.; Ren, J.; Hillier, J.; Lu, W.; Jones, E.Y. Antiepileptic Drug Carbamazepine Binds to a Novel Pocket on the Wnt Receptor Frizzled-8. J. Med. Chem. 2020, 63, 3252–3260. [Google Scholar] [CrossRef] [PubMed]
  445. Fujii, N.; You, L.; Xu, Z.; Uematsu, K.; Shan, J.; He, B.; Mikami, I.; Edmondson, L.R.; Neale, G.; Zheng, J.; et al. An Antagonist of Dishevelled Protein-Protein Interaction Suppresses β-Catenin–Dependent Tumor Cell Growth. Cancer Res. 2007, 67, 573–579. [Google Scholar] [CrossRef]
  446. Nile, A.H.; De Sousa E Melo, F.; Mukund, S.; Piskol, R.; Hansen, S.; Zhou, L.; Zhang, Y.; Fu, Y.; Gogol, E.B.; Kömüves, L.G.; et al. A Selective Peptide Inhibitor of Frizzled 7 Receptors Disrupts Intestinal Stem Cells. Nat. Chem. Biol. 2018, 14, 582–590. [Google Scholar] [CrossRef] [PubMed]
  447. Osada, T.; Chen, M.; Yang, X.Y.; Spasojevic, I.; Vandeusen, J.B.; Hsu, D.; Clary, B.M.; Clay, T.M.; Chen, W.; Morse, M.A.; et al. Antihelminth Compound Niclosamide Downregulates Wnt Signaling and Elicits Antitumor Responses in Tumors with Activating APC Mutations. Cancer Res. 2011, 71, 4172–4182. [Google Scholar] [CrossRef] [PubMed]
  448. Lin, C.K.; Bai, M.Y.; Hu, T.M.; Wang, Y.C.; Chao, T.K.; Weng, S.J.; Huang, R.L.; Su, P.H.; Lai, H.C. Preclinical Evaluation of a Nanoformulated Antihelminthic, Niclosamide, in Ovarian Cancer. Oncotarget 2016, 7, 8993. [Google Scholar] [CrossRef]
  449. Schweizer, M.T.; Haugk, K.; McKiernan, J.S.; Gulati, R.; Cheng, H.H.; Maes, J.L.; Dumpit, R.F.; Nelson, P.S.; Montgomery, B.; McCune, J.S.; et al. A Phase I Study of Niclosamide in Combination with Enzalutamide in Men with Castration-Resistant Prostate Cancer. PLoS ONE 2018, 13, e0198389. [Google Scholar] [CrossRef]
  450. Chen, B.; Dodge, M.E.; Tang, W.; Lu, J.; Ma, Z.; Fan, C.W.; Wei, S.; Hao, W.; Kilgore, J.; Williams, N.S.; et al. Small Molecule–Mediated Disruption of Wnt-Dependent Signaling in Tissue Regeneration and Cancer. Nat. Chem. Biol. 2009, 5, 100–107. [Google Scholar] [CrossRef] [PubMed]
  451. Ko, A.H.; Chiorean, E.G.; Kwak, E.L.; Lenz, H.-J.; Nadler, P.I.; Wood, D.L.; Fujimori, M.; Inada, T.; Kouji, H.; McWilliams, R.R. Final Results of a Phase Ib Dose-Escalation Study of PRI-724, a CBP/Beta-Catenin Modulator, plus Gemcitabine (GEM) in Patients with Advanced Pancreatic Adenocarcinoma (APC) as Second-Line Therapy after FOLFIRINOX or FOLFOX. J. Clin. Oncol. 2016, 34 (Suppl. S15), e15721. [Google Scholar] [CrossRef]
  452. El-Khoueiry, A.B.; Ning, Y.; Yang, D.; Cole, S.; Kahn, M.; Zoghbi, M.; Berg, J.; Fujimori, M.; Inada, T.; Kouji, H.; et al. A Phase I First-in-Human Study of PRI-724 in Patients (Pts) with Advanced Solid Tumors. J. Clin. Oncol. 2013, 31 (Suppl. S15), 2501. [Google Scholar] [CrossRef]
  453. Ben-Porath, I.; Thomson, M.W.; Carey, V.J.; Ge, R.; Bell, G.W.; Regev, A.; Weinberg, R.A. An Embryonic Stem Cell–like Gene Expression Signature in Poorly Differentiated Aggressive Human Tumors. Nat. Genet. 2008, 40, 499–507. [Google Scholar] [CrossRef] [PubMed]
  454. Schaefer, T.; Lengerke, C. SOX2 Protein Biochemistry in Stemness, Reprogramming, and Cancer: The PI3K/AKT/SOX2 Axis and Beyond. Oncogene 2019, 39, 278–292. [Google Scholar] [CrossRef] [PubMed]
  455. Avilion, A.A.; Nicolis, S.K.; Pevny, L.H.; Perez, L.; Vivian, N.; Lovell-Badge, R. Multipotent Cell Lineages in Early Mouse Development Depend on SOX2 Function. Genes Dev. 2003, 17, 126–140. [Google Scholar] [CrossRef] [PubMed]
  456. Takahashi, K.; Yamanaka, S. Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed]
  457. Arnold, K.; Sarkar, A.; Yram, M.A.; Polo, J.M.; Bronson, R.; Sengupta, S.; Seandel, M.; Geijsen, N.; Hochedlinger, K. Sox2(+) Adult Stem and Progenitor Cells Are Important for Tissue Regeneration and Survival of Mice. Cell Stem Cell 2011, 9, 317–329. [Google Scholar] [CrossRef] [PubMed]
  458. Liu, K.; Xie, F.; Zhao, T.; Zhang, R.; Gao, A.; Chen, Y.; Li, H.; Zhang, S.; Xiao, Z.; Li, J.; et al. Targeting SOX2 Protein with Peptide Aptamers for Therapeutic Gains against Esophageal Squamous Cell Carcinoma. Mol. Ther. 2020, 28, 901–913. [Google Scholar] [CrossRef]
  459. Al Mamun, M.; Mannoor, K.; Cao, J.; Qadri, F.; Song, X. SOX2 in Cancer Stemness: Tumor Malignancy and Therapeutic Potentials. J. Mol. Cell Biol. 2020, 12, 85. [Google Scholar] [CrossRef]
  460. Zhang, S.; Xiong, X.; Sun, Y. Functional Characterization of SOX2 as an Anticancer Target. Signal Transduct. Target. Ther. 2020, 5, 135. [Google Scholar] [CrossRef] [PubMed]
  461. Wang, K.; Ji, W.; Yu, Y.; Li, Z.; Niu, X.; Xia, W.; Lu, S. FGFR1-ERK1/2-SOX2 Axis Promotes Cell Proliferation, Epithelial-Mesenchymal Transition, and Metastasis in FGFR1-Amplified Lung Cancer. Oncogene 2018, 37, 5340–5354. [Google Scholar] [CrossRef] [PubMed]
  462. Ji, J.; Yu, Y.; Li, Z.L.; Chen, M.Y.; Deng, R.; Huang, X.; Wang, G.F.; Zhang, M.X.; Yang, Q.; Ravichandran, S.; et al. XIAP Limits Autophagic Degradation of Sox2 and Is A Therapeutic Target in Nasopharyngeal Carcinoma Stem Cells. Theranostics 2018, 8, 1494. [Google Scholar] [CrossRef]
  463. Boumahdi, S.; Driessens, G.; Lapouge, G.; Rorive, S.; Nassar, D.; Le Mercier, M.; Delatte, B.; Caauwe, A.; Lenglez, S.; Nkusi, E.; et al. SOX2 Controls Tumor Initiation and Cancer Stem-Cell Functions in Squamous-Cell Carcinoma. Nature 2014, 511, 246–250. [Google Scholar] [CrossRef] [PubMed]
  464. Zhu, Y.; Huang, S.; Chen, S.; Chen, J.; Wang, Z.; Wang, Y.; Zheng, H. SOX2 Promotes Chemoresistance, Cancer Stem Cells Properties, and Epithelial–Mesenchymal Transition by β-Catenin and Beclin1/Autophagy Signaling in Colorectal Cancer. Cell Death Dis. 2021, 12, 449. [Google Scholar] [CrossRef] [PubMed]
  465. Stolzenburg, S.; Rots, M.G.; Beltran, A.S.; Rivenbark, A.G.; Yuan, X.; Qian, H.; Strahl, B.D.; Blancafort, P. Targeted Silencing of the Oncogenic Transcription Factor SOX2 in Breast Cancer. Nucleic Acids Res. 2012, 40, 6725. [Google Scholar] [CrossRef] [PubMed]
  466. Yokota, E.; Yamatsuji, T.; Takaoka, M.; Haisa, M.; Takigawa, N.; Miyake, N.; Ikeda, T.; Mori, T.; Ohno, S.; Sera, T.; et al. Targeted Silencing of SOX2 by an Artificial Transcription Factor Showed Antitumor Effect in Lung and Esophageal Squamous Cell Carcinoma. Oncotarget 2017, 8, 103063–103076. [Google Scholar] [CrossRef]
  467. Taniguchi, J.; Pandian, G.N.; Hidaka, T.; Hashiya, K.; Bando, T.; Kim, K.K.; Sugiyama, H. A Synthetic DNA-Binding Inhibitor of SOX2 Guides Human Induced Pluripotent Stem Cells to Differentiate into Mesoderm. Nucleic Acids Res. 2017, 45, 9219. [Google Scholar] [CrossRef] [PubMed]
  468. Gandhi, N.S.; Wang, E.; Sorolla, A.; Kan, Y.J.; Malik, A.; Batra, J.; Young, K.A.; Tie, W.J.; Blancafort, P.; Mancera, R.L. Design and Characterization of a Cell-Penetrating Peptide Derived from the Sox2 Transcription Factor. Int. J. Mol. Sci. 2021, 22, 9354. [Google Scholar] [CrossRef]
  469. Schmitz, M.; Temme, A.; Senner, V.; Ebner, R.; Schwind, S.; Stevanovic, S.; Wehner, R.; Schackert, G.; Schackert, H.K.; Fussel, M.; et al. Identification of SOX2 as a Novel Glioma-Associated Antigen and Potential Target for T Cell-Based Immunotherapy. Br. J. Cancer 2007, 96, 1293–1301. [Google Scholar] [CrossRef]
  470. Favaro, R.; Appolloni, I.; Pellegatta, S.; Sanga, A.B.; Pagella, P.; Gambini, E.; Pisati, F.; Ottolenghi, S.; Foti, M.; Finocchiaro, G.; et al. Sox2 Is Required to Maintain Cancer Stem Cells in a Mouse Model of High-Grade Oligodendroglioma. Cancer Res. 2014, 74, 1833–1844. [Google Scholar] [CrossRef] [PubMed]
  471. Garros-Regulez, L.; Garcia, I.; Carrasco-Garcia, E.; Lantero, A.; Aldaz, P.; Moreno-Cugnon, L.; Arrizabalaga, O.; Undabeitia, J.; Torres-Bayona, S.; Villanua, J.; et al. Targeting SOX2 as a Therapeutic Strategy in Glioblastoma. Front. Oncol. 2016, 6, 225912. [Google Scholar] [CrossRef]
  472. Mohamed, M.A.; Elkhateeb, W.A.; Daba, G.M. Rapamycin Golden Jubilee and Still the Miraculous Drug: A Potent Immunosuppressant, Antitumor, Rejuvenative Agent, and Potential Contributor in COVID-19 Treatment. Bioresour. Bioprocess 2022, 9, 65. [Google Scholar] [CrossRef] [PubMed]
  473. Garros-Regulez, L.; Aldaz, P.; Arrizabalaga, O.; Moncho-Amor, V.; Carrasco-Garcia, E.; Manterola, L.; Moreno-Cugnon, L.; Barrena, C.; Villanua, J.; Ruiz, I.; et al. MTOR Inhibition Decreases SOX2-SOX9 Mediated Glioma Stem Cell Activity and Temozolomide Resistance. Expert Opin. Ther. Targets 2016, 20, 393–405. [Google Scholar] [CrossRef]
  474. Wang, Z.; Kang, L.; Zhang, H.; Huang, Y.; Fang, L.; Li, M.; Brown, P.J.; Arrowsmith, C.H.; Li, J.; Wong, J. AKT Drives SOX2 Overexpression and Cancer Cell Stemness in Esophageal Cancer by Protecting SOX2 from UBR5-Mediated Degradation. Oncogene 2019, 38, 5250–5264. [Google Scholar] [CrossRef] [PubMed]
  475. Xing, Y.; Lin, N.U.; Maurer, M.A.; Chen, H.; Mahvash, A.; Sahin, A.; Akcakanat, A.; Li, Y.; Abramson, V.; Litton, J.; et al. Phase II Trial of AKT Inhibitor MK-2206 in Patients with Advanced Breast Cancer Who Have Tumors with PIK3CA or AKT Mutations, and/or PTEN Loss/PTEN Mutation. Breast Cancer Res. 2019, 21, 78. [Google Scholar] [CrossRef] [PubMed]
  476. Qin, J.; Ji, J.; Deng, R.; Tang, J.; Yang, F.; Feng, G.K.; Chen, W.D.; Wu, X.Q.; Qian, X.J.; Ding, K.; et al. DC120, a Novel AKT Inhibitor, Preferentially Suppresses Nasopharyngeal Carcinoma Cancer Stem-like Cells by Downregulating Sox2. Oncotarget 2015, 6, 6944. [Google Scholar] [CrossRef] [PubMed]
  477. Coombes, R.C.; Badman, P.D.; Lozano-Kuehne, J.P.; Liu, X.; Macpherson, I.R.; Zubairi, I.; Baird, R.D.; Rosenfeld, N.; Garcia-Corbacho, J.; Cresti, N.; et al. Results of the Phase IIa RADICAL Trial of the FGFR Inhibitor AZD4547 in Endocrine Resistant Breast Cancer. Nat. Commun. 2022, 13, 3246. [Google Scholar] [CrossRef] [PubMed]
  478. Singh, S.; Trevino, J.; Bora-Singhal, N.; Coppola, D.; Haura, E.; Altiok, S.; Chellappan, S.P. EGFR/Src/Akt Signaling Modulates Sox2 Expression and Self-Renewal of Stem-like Side-Population Cells in Non-Small Cell Lung Cancer. Mol. Cancer 2012, 11, 73. [Google Scholar] [CrossRef] [PubMed]
  479. Pietrobono, S.; Morandi, A.; Gagliardi, S.; Gerlini, G.; Borgognoni, L.; Chiarugi, P.; Arbiser, J.L.; Stecca, B. Down-Regulation of SOX2 Underlies the Inhibitory Effects of the Triphenylmethane Gentian Violet on Melanoma Cell Self-Renewal and Survival. J. Investig. Dermatol. 2016, 136, 2059–2069. [Google Scholar] [CrossRef]
  480. Kim, B.R.; Coyaud, E.; Laurent, E.M.N.; St-Germain, J.; Van De Laar, E.; Tsao, M.S.; Raught, B.; Moghal, N. Identification of the SOX2 Interactome by BioID Reveals EP300 as a Mediator of SOX2-Dependent Squamous Differentiation and Lung Squamous Cell Carcinoma Growth. Mol. Cell. Proteom. 2017, 16, 1864–1888. [Google Scholar] [CrossRef] [PubMed]
  481. Yin, Y.; Xie, C.M.; Li, H.; Tan, M.; Chen, G.; Schiff, R.; Xiong, X.; Sun, Y. The FBXW2–MSX2–SOX2 Axis Regulates Stem Cell Property and Drug Resistance of Cancer Cells. Proc. Natl. Acad. Sci. USA 2019, 116, 20528–20538. [Google Scholar] [CrossRef] [PubMed]
  482. Swords, R.T.; Erba, H.P.; DeAngelo, D.J.; Smith, P.G.; Pickard, M.D.; Dezube, B.J.; Giles, F.J.; Medeiros, B.C. The Novel, Investigational NEDD8-Activating Enzyme Inhibitor MLN4924 In Adult Patients with Acute Myeloid Leukemia (AML) or High-Grade Myelodysplastic Syndromes (MDS): A Phase 1 Study. Blood 2010, 116, 658. [Google Scholar] [CrossRef]
  483. Nawrocki, S.T.; Griffin, P.; Kelly, K.R.; Carew, J.S. MLN4924: A Novel First-in-Class Inhibitor of NEDD8-Activating Enzyme for Cancer Therapy. Expert Opin. Investig. Drugs 2012, 21, 1563–1573. [Google Scholar] [CrossRef] [PubMed]
  484. Pan, W.; Luo, Q.; Yan, X.; Yuan, L.; Yi, H.; Zhang, L.; Li, B.; Zhang, Y.; Sun, J.; Qiu, M.Z.; et al. A Novel SMAC Mimetic APG-1387 Exhibits Dual Antitumor Effect on HBV-Positive Hepatocellular Carcinoma with High Expression of CIAP2 by Inducing Apoptosis and Enhancing Innate Anti-Tumor Immunity. Biochem. Pharmacol. 2018, 154, 127–135. [Google Scholar] [CrossRef]
  485. Bora-Singhal, N.; Mohankumar, D.; Saha, B.; Colin, C.M.; Lee, J.Y.; Martin, M.W.; Zheng, X.; Coppola, D.; Chellappan, S. Novel HDAC11 Inhibitors Suppress Lung Adenocarcinoma Stem Cell Self-Renewal and Overcome Drug Resistance by Suppressing Sox2. Sci. Rep. 2020, 10, 4722. [Google Scholar] [CrossRef] [PubMed]
  486. Zhang, X.; Lu, F.; Wang, J.; Yin, F.; Xu, Z.; Qi, D.; Wu, X.; Cao, Y.; Liang, W.; Liu, Y.; et al. Pluripotent Stem Cell Protein Sox2 Confers Sensitivity to LSD1 Inhibition in Cancer Cells. Cell Rep. 2013, 5, 445–457. [Google Scholar] [CrossRef]
  487. Cuyàs, E.; Gumuzio, J.; Verdura, S.; Brunet, J.; Bosch-Barrera, J.; Martin-Castillo, B.; Alarcón, T.; Encinar, J.A.; Martin, Á.G.; Menendez, J.A. The LSD1 Inhibitor Iadademstat (ORY-1001) Targets SOX2-Driven Breast Cancer Stem Cells: A Potential Epigenetic Therapy in Luminal-B and HER2-Positive Breast Cancer Subtypes. Aging 2020, 12, 4794. [Google Scholar] [CrossRef]
  488. Somervaille, T.; Salamero, O.; Montesinos, P.; Willekens, C.; Perez Simon, J.A.; Pigneux, A.; Recher, C.; Popat, R.; Molinero, C.; Mascaro, C.; et al. Safety, Phamacokinetics (PK), Pharmacodynamics (PD) and Preliminary Activity in Acute Leukemia of Ory-1001, a First-in-Class Inhibitor of Lysine-Specific Histone Demethylase 1A (LSD1/KDM1A): Initial Results from a First-in-Human Phase 1 Study. Blood 2016, 128, 4060. [Google Scholar] [CrossRef]
  489. Cochrane, C.R.; Szczepny, A.; Watkins, D.N.; Cain, J.E. Hedgehog Signaling in the Maintenance of Cancer Stem Cells. Cancers 2015, 7, 1554–1585. [Google Scholar] [CrossRef]
  490. Sari, I.N.; Phi, L.T.H.; Jun, N.; Wijaya, Y.T.; Lee, S.; Kwon, H.Y. Hedgehog Signaling in Cancer: A Prospective Therapeutic Target for Eradicating Cancer Stem Cells. Cells 2018, 7, 208. [Google Scholar] [CrossRef]
  491. Liu, S.; Dontu, G.; Mantle, I.D.; Patel, S.; Ahn, N.S.; Jackson, K.W.; Suri, P.; Wicha, M.S. Hedgehog Signaling and Bmi-1 Regulate Self-Renewal of Normal and Malignant Human Mammary Stem Cells. Cancer Res. 2006, 66, 6063–6071. [Google Scholar] [CrossRef]
  492. Santini, R.; Vinci, M.C.; Pandolfi, S.; Penachioni, J.Y.; Montagnani, V.; Olivito, B.; Gattai, R.; Pimpinelli, N.; Gerlini, G.; Borgognoni, L.; et al. HEDGEHOG-GLI Signaling Drives Self-Renewal and Tumorigenicity of Human Melanoma-Initiating Cells. Stem Cells 2012, 30, 1808–1818. [Google Scholar] [CrossRef]
  493. Zhao, C.; Chen, A.; Jamieson, C.H.; Fereshteh, M.; Abrahamsson, A.; Blum, J.; Kwon, H.Y.; Kim, J.; Chute, J.P.; Rizzieri, D.; et al. Hedgehog Signaling Is Essential for Maintenance of Cancer Stem Cells in Myeloid Leukaemia. Nature 2009, 458, 776–779. [Google Scholar] [CrossRef] [PubMed]
  494. Miyazaki, Y.; Matsubara, S.; Ding, Q.; Tsukasa, K.; Yoshimitsu, M.; Kosai, K.I.; Takao, S. Efficient Elimination of Pancreatic Cancer Stem Cells by Hedgehog/GLI Inhibitor GANT61 in Combination with MTOR Inhibition. Mol. Cancer 2016, 15, 49. [Google Scholar] [CrossRef]
  495. Carpenter, R.L.; Ray, H. Efficacy and Safety of Sonic Hedgehog Pathway Inhibitors in Cancer. Drug Saf. 2019, 42, 263. [Google Scholar] [CrossRef]
  496. Meiss, F.; Andrlová, H.; Zeiser, R. Vismodegib. Recent. Results Cancer Res. 2018, 211, 125–139. [Google Scholar] [CrossRef] [PubMed]
  497. Singh, B.N.; Fu, J.; Srivastava, R.K.; Shankar, S. Hedgehog Signaling Antagonist GDC-0449 (Vismodegib) Inhibits Pancreatic Cancer Stem Cell Characteristics: Molecular Mechanisms. PLoS ONE 2011, 6, e27306. [Google Scholar] [CrossRef] [PubMed]
  498. Sekulic, A.; Migden, M.R.; Basset-Seguin, N.; Garbe, C.; Gesierich, A.; Lao, C.D.; Miller, C.; Mortier, L.; Murrell, D.F.; Hamid, O.; et al. Long-Term Safety and Efficacy of Vismodegib in Patients with Advanced Basal Cell Carcinoma: Final Update of the Pivotal ERIVANCE BCC Study. BMC Cancer 2017, 17, 332. [Google Scholar] [CrossRef]
  499. Von Hoff, D.D.; LoRusso, P.M.; Rudin, C.M.; Reddy, J.C.; Yauch, R.L.; Tibes, R.; Weiss, G.J.; Borad, M.J.; Hann, C.L.; Brahmer, J.R.; et al. Inhibition of the Hedgehog Pathway in Advanced Basal-Cell Carcinoma. N. Engl. J. Med. 2009, 361, 1164–1172. [Google Scholar] [CrossRef]
  500. Kim, R.; Ji, J.H.; Kim, J.H.; Hong, J.Y.; Lim, H.Y.; Kang, W.K.; Lee, J.; Kim, S.T. Safety and Anti-Tumor Effects of Vismodegib in Patients with Refractory Advanced Gastric Cancer: A Single-Arm, Phase-II Trial. J. Cancer 2022, 13, 1097–1102. [Google Scholar] [CrossRef] [PubMed]
  501. Kaye, S.B.; Fehrenbacher, L.; Holloway, R.; Amit, A.; Karlan, B.; Slomovitz, B.; Sabbatini, P.; Fu, L.; Yauch, R.L.; Chang, I.; et al. A Phase II, Randomized, Placebo-Controlled Study of Vismodegib as Maintenance Therapy in Patients with Ovarian Cancer in Second or Third Complete Remission. Clin. Cancer Res. 2012, 18, 6509–6518. [Google Scholar] [CrossRef]
  502. De Jesus-Acosta, A.; Sugar, E.A.; O’Dwyer, P.J.; Ramanathan, R.K.; Von Hoff, D.D.; Rasheed, Z.; Zheng, L.; Begum, A.; Anders, R.; Maitra, A.; et al. Phase 2 Study of Vismodegib, a Hedgehog Inhibitor, Combined with Gemcitabine and Nab-Paclitaxel in Patients with Untreated Metastatic Pancreatic Adenocarcinoma. Br. J. Cancer 2020, 122, 498–505. [Google Scholar] [CrossRef]
  503. Burness, C.B. Sonidegib: First Global Approval. Drugs 2015, 75, 1559–1566. [Google Scholar] [CrossRef]
  504. Irvine, D.A.; Zhang, B.; Kinstrie, R.; Tarafdar, A.; Morrison, H.; Campbell, V.L.; Moka, H.A.; Ho, Y.; Nixon, C.; Manley, P.W.; et al. Deregulated Hedgehog Pathway Signaling Is Inhibited by the Smoothened Antagonist LDE225 (Sonidegib) in Chronic Phase Chronic Myeloid Leukaemia. Sci. Rep. 2016, 6, 25476. [Google Scholar] [CrossRef] [PubMed]
  505. Zhang, Y.; Vagiannis, D.; Budagaga, Y.; Sabet, Z.; Hanke, I.; Rozkoš, T.; Hofman, J. Sonidegib Potentiates the Cancer Cells’ Sensitivity to Cytostatic Agents by Functional Inhibition of ABCB1 and ABCG2 in Vitro and Ex Vivo. Biochem. Pharmacol. 2022, 199, 115009. [Google Scholar] [CrossRef]
  506. Dummer, R.; Guminksi, A.; Gutzmer, R.; Lear, J.T.; Lewis, K.D.; Chang, A.L.S.; Combemale, P.; Dirix, L.; Kaatz, M.; Kudchadkar, R.; et al. Long-term Efficacy and Safety of Sonidegib in Patients with Advanced Basal Cell Carcinoma: 42-month Analysis of the Phase II Randomized, Double-blind BOLT Study. Br. J. Dermatol. 2020, 182, 1369–1378. [Google Scholar] [CrossRef]
  507. Pietanza, M.C.; Litvak, A.M.; Varghese, A.M.; Krug, L.M.; Fleisher, M.; Teitcher, J.B.; Holodny, A.I.; Sima, C.S.; Woo, K.M.; Ng, K.K.; et al. A Phase I Trial of the Hedgehog Inhibitor, Sonidegib (LDE225), in Combination with Etoposide and Cisplatin for the Initial Treatment of Extensive Stage Small Cell Lung Cancer. Lung Cancer 2016, 99, 23–30. [Google Scholar] [CrossRef] [PubMed]
  508. Cazet, A.S.; Hui, M.N.; Elsworth, B.L.; Wu, S.Z.; Roden, D.; Chan, C.L.; Skhinas, J.N.; Collot, R.; Yang, J.; Harvey, K.; et al. Targeting Stromal Remodeling and Cancer Stem Cell Plasticity Overcomes Chemoresistance in Triple Negative Breast Cancer. Nat. Commun. 2018, 9, 2897. [Google Scholar] [CrossRef]
  509. Munchhof, M.J.; Li, Q.; Shavnya, A.; Borzillo, G.V.; Boyden, T.L.; Jones, C.S.; Lagreca, S.D.; Martinez-Alsina, L.; Patel, N.; Pelletier, K.; et al. Discovery of PF-04449913, a Potent and Orally Bioavailable Inhibitor of Smoothened. ACS Med. Chem. Lett. 2011, 3, 106–111. [Google Scholar] [CrossRef]
  510. Hoy, S.M. Glasdegib: First Global Approval. Drugs 2019, 79, 207–213. [Google Scholar] [CrossRef] [PubMed]
  511. Cortes, J.E.; Heidel, F.H.; Fiedler, W.; Smith, B.D.; Robak, T.; Montesinos, P.; Candoni, A.; Leber, B.; Sekeres, M.A.; Pollyea, D.A.; et al. Survival Outcomes and Clinical Benefit in Patients with Acute Myeloid Leukemia Treated with Glasdegib and Low-Dose Cytarabine According to Response to Therapy. J. Hematol. Oncol. 2020, 13, 92. [Google Scholar] [CrossRef] [PubMed]
  512. Sekeres, M.A.; Montesinos, P.; Novak, J.; Wang, J.; Jeyakumar, D.; Tomlinson, B.; Mayer, J.; Jou, E.; Robak, T.; Taussig, D.C.; et al. Glasdegib plus Intensive or Non-Intensive Chemotherapy for Untreated Acute Myeloid Leukemia: Results from the Randomized, Phase 3 BRIGHT AML 1019 Trial. Leukemia 2023, 37, 2017–2026. [Google Scholar] [CrossRef] [PubMed]
  513. Chen, J.K.; Taipale, J.; Cooper, M.K.; Beachy, P.A. Inhibition of Hedgehog Signaling by Direct Binding of Cyclopamine to Smoothened. Genes Dev. 2002, 16, 2743. [Google Scholar] [CrossRef]
  514. Che, J.; Zhang, F.Z.; Zhao, C.Q.; Hu, X.D.; Fan, S.J. Cyclopamine Is a Novel Hedgehog Signaling Inhibitor with Significant Anti-Proliferative, Anti-Invasive and Anti-Estrogenic Potency in Human Breast Cancer Cells. Oncol. Lett. 2013, 5, 1417. [Google Scholar] [CrossRef]
  515. Feldmann, G.; Fendrich, V.; McGovern, K.; Bedja, D.; Bisht, S.; Alvarez, H.; Koorstra, J.B.M.; Habbe, N.; Karikari, C.; Mullendore, M.; et al. An Orally Bioavailable Small-Molecule Inhibitor of Hedgehog Signaling Inhibits Tumor Initiation and Metastasis in Pancreatic Cancer. Mol. Cancer Ther. 2008, 7, 2725. [Google Scholar] [CrossRef] [PubMed]
  516. Tremblay, M.R.; Lescarbeau, A.; Grogan, M.J.; Tan, E.; Lin, G.; Austad, B.C.; Yu, L.C.; Behnke, M.L.; Nair, S.J.; Hagel, M.; et al. Discovery of a Potent and Orally Active Hedgehog Pathway Antagonist (IPI-926). J. Med. Chem. 2009, 52, 4400–4418. [Google Scholar] [CrossRef]
  517. Campbell, V.T.; Nadesan, P.; Ali, S.A.; Wang, C.Y.Y.; Whetstone, H.; Poon, R.; Wei, Q.; Keilty, J.; Proctor, J.; Wang, L.W.; et al. Hedgehog Pathway Inhibition in Chondrosarcoma Using the Smoothened Inhibitor IPI-926 Directly Inhibits Sarcoma Cell Growth. Mol. Cancer Ther. 2014, 13, 1259–1269. [Google Scholar] [CrossRef] [PubMed]
  518. Bowles, D.W.; Keysar, S.B.; Eagles, J.R.; Wang, G.; Glogowska, M.J.; McDermott, J.D.; Le, P.N.; Gao, D.; Ray, C.E.; Rochon, P.J.; et al. A Pilot Study of Cetuximab and the Hedgehog Inhibitor IPI-926 in Recurrent/Metastatic Head and Neck Squamous Cell Carcinoma. Oral. Oncol. 2016, 53, 74. [Google Scholar] [CrossRef]
  519. Jimeno, A.; Weiss, G.J.; Miller, W.H.; Gettinger, S.; Eigl, B.J.C.; Chang, A.L.S.; Dunbar, J.; Devens, S.; Faia, K.; Skliris, G.; et al. Phase I Study of the Hedgehog Pathway Inhibitor IPI-926 in Adult Patients with Solid Tumors. Clin. Cancer Res. 2013, 19, 2766–2774. [Google Scholar] [CrossRef]
  520. Ko, A.H.; LoConte, N.; Tempero, M.A.; Walker, E.J.; Kelley, R.K.; Lewis, S.; Chang, W.C.; Kantoff, E.; Vannier, M.W.; Catenacci, D.V.; et al. A Phase I Study of FOLFIRINOX Plus IPI-926, a Hedgehog Pathway Inhibitor, for Advanced Pancreatic Adenocarcinoma. Pancreas 2016, 45, 370. [Google Scholar] [CrossRef] [PubMed]
  521. van Maldegem, A.M.; Bovée, J.V.; Gelderblom, H. Comprehensive Analysis of Published Studies Involving Systemic Treatment for Chondrosarcoma of Bone between 2000 and 2013. Clin. Sarcoma Res. 2014, 4, 11. [Google Scholar] [CrossRef] [PubMed]
  522. Zaidi, A.H.; Komatsu, Y.; Kelly, L.A.; Malhotra, U.; Rotoloni, C.; Kosovec, J.E.; Zahoor, H.; Makielski, R.; Bhatt, A.; Hoppo, T.; et al. Smoothened Inhibition Leads to Decreased Proliferation and Induces Apoptosis in Esophageal Adenocarcinoma Cells. Cancer Investig. 2013, 31, 480–489. [Google Scholar] [CrossRef] [PubMed]
  523. Rocconi, R.; Meng, E.; Shevde, L.; Finan, M.; Reed, E. A Novel Hedgehog Pathway Smoothened Inhibitor (BMS-833923) Demonstrates in Vitro Synergy with Carboplatin in Ovarian Cancer Cells. Gynecol. Oncol. 2011, 120, S41–S42. [Google Scholar] [CrossRef]
  524. Arenas, A.; Lainez, D.; Serrano del Castillo, C.; Salgado, R.N.; Blas, C.; Atance, M.; Cordoba, R.; Llamas, P.; Alonso Dominguez, J.M. Inhibition of the Hedgehog Pathway Decreases the Quiescent CD34+CD38- Population in Acute Myeloid Leukemia. Blood 2018, 132 (Suppl. S1), 1509. [Google Scholar] [CrossRef]
  525. Shah, N.P.; Cortes, J.E.; Martinelli, G.; Smith, B.D.; Clarke, E.; Copland, M.; Strauss, L.; Talpaz, M. Dasatinib Plus Smoothened (SMO) Inhibitor BMS-833923 in Chronic Myeloid Leukemia (CML) with Resistance or Suboptimal Response to a Prior Tyrosine Kinase Inhibitor (TKI): Phase I Study CA180323. Blood 2014, 124, 4539. [Google Scholar] [CrossRef]
  526. Bender, M.H.; Hipskind, P.A.; Capen, A.R.; Cockman, M.; Credille, K.M.; Gao, H.; Bastian, J.A.; Clay, J.M.; Lobb, K.L.; Sall, D.J.; et al. Abstract 2819: Identification and Characterization of a Novel Smoothened Antagonist for the Treatment of Cancer with Deregulated Hedgehog Signaling. Cancer Res. 2011, 71 (Suppl. S8), 2819. [Google Scholar] [CrossRef]
  527. Bendell, J.; Andre, V.; Ho, A.; Kudchadkar, R.; Migden, M.; Infante, J.; Tiu, R.V.; Pitou, C.; Tucker, T.; Brail, L.; et al. Phase i Study of Ly2940680, a Smo Antagonist, in Patients with Advanced Cancer Including Treatment-Naïve and Previously Treated Basal Cell Carcinoma. Clin. Cancer Res. 2018, 24, 2082–2091. [Google Scholar] [CrossRef] [PubMed]
  528. Chen, Q.; Zhang, H.; Wu, M.; Wang, Q.; Luo, L.; Ma, H.; Zhang, X.; He, S. Discovery of a Potent Hedgehog Pathway Inhibitor Capable of Activating Caspase8-Dependent Apoptosis. J. Pharmacol. Sci. 2018, 137, 256–264. [Google Scholar] [CrossRef]
  529. Li, C.L.; Fang, Z.X.; Wu, Z.; Hou, Y.Y.; Wu, H.T.; Liu, J. Repurposed Itraconazole for Use in the Treatment of Malignancies as a Promising Therapeutic Strategy. Biomed. Pharmacother. 2022, 154, 113616. [Google Scholar] [CrossRef]
  530. Kim, J.; Tang, J.Y.; Gong, R.; Kim, J.; Lee, J.J.; Clemons, K.V.; Chong, C.R.; Chang, K.S.; Fereshteh, M.; Gardner, D.; et al. Itraconazole, a Commonly Used Anti-Fungal That Inhibits Hedgehog Pathway Activity and Cancer Growth. Cancer Cell 2010, 17, 388. [Google Scholar] [CrossRef] [PubMed]
  531. Deng, H.; Huang, L.; Liao, Z.; Liu, M.; Li, Q.; Xu, R. Itraconazole Inhibits the Hedgehog Signaling Pathway Thereby Inducing Autophagy-Mediated Apoptosis of Colon Cancer Cells. Cell Death Dis. 2020, 11, 539. [Google Scholar] [CrossRef] [PubMed]
  532. Wang, X.; Wei, S.; Zhao, Y.; Shi, C.; Liu, P.; Zhang, C.; Lei, Y.; Zhang, B.; Bai, B.; Huang, Y.; et al. Anti-Proliferation of Breast Cancer Cells with Itraconazole: Hedgehog Pathway Inhibition Induces Apoptosis and Autophagic Cell Death. Cancer Lett. 2017, 385, 128–136. [Google Scholar] [CrossRef] [PubMed]
  533. Tsubamoto, H.; Sonoda, T.; Inoue, K. Impact of Itraconazole on the Survival of Heavily Pre-Treated Patients with Triple-Negative Breast Cancer. Anticancer Res. 2014, 34, 3839–3844. [Google Scholar]
  534. Inoue, K.; Tsubamoto, H.; Sonoda, T.; Ikuta, S.; Tani, S.; Yamanaka, N. Efficacy of Combination Chemotherapy with Itraconazole for Treating Metastatic Pancreatic Cancer in the Second-Line Setting. J. Clin. Oncol. 2015, 33 (Suppl. S15), e15255. [Google Scholar] [CrossRef]
  535. Tsubamoto, H.; Sonoda, T.; Inoue, K.; Ikuta, S.; Tani, S.; Yamanaka, N. Impact of Itraconazole after First-Line Chemotherapy on the Survival of Patients with Distant Metastatic Biliary Tract Cancer. J. Clin. Oncol. 2015, 33 (Suppl. S15), e15145. [Google Scholar] [CrossRef]
  536. Rudin, C.M.; Brahmer, J.R.; Juergens, R.A.; Hann, C.L.; Ettinger, D.S.; Sebree, R.; Smith, R.; Aftab, B.T.; Huang, P.; Liu, J.O. Phase 2 Study of Pemetrexed and Itraconazole as Second-Line Therapy for Metastatic Nonsquamous Non–Small-Cell Lung Cancer. J. Thorac. Oncol. 2013, 8, 619–623. [Google Scholar] [CrossRef] [PubMed]
  537. Antonarakis, E.S.; Heath, E.I.; Smith, D.C.; Rathkopf, D.; Blackford, A.L.; Danila, D.C.; King, S.; Frost, A.; Ajiboye, A.S.; Zhao, M.; et al. Repurposing Itraconazole as a Treatment for Advanced Prostate Cancer: A Noncomparative Randomized Phase II Trial in Men with Metastatic Castration-Resistant Prostate Cancer. Oncologist 2013, 18, 163–173. [Google Scholar] [CrossRef]
  538. Kim, J.; Lee, J.J.; Kim, J.; Gardner, D.; Beachy, P.A. Arsenic Antagonizes the Hedgehog Pathway by Preventing Ciliary Accumulation and Reducing Stability of the Gli2 Transcriptional Effector. Proc. Natl. Acad. Sci. USA 2010, 107, 13432–13437. [Google Scholar] [CrossRef]
  539. Yang, D.; Cao, F.; Ye, X.; Zhao, H.; Liu, X.; Li, Y.; Shi, C.; Wang, H.; Zhou, J. Arsenic Trioxide Inhibits the Hedgehog Pathway Which Is Aberrantly Activated in Acute Promyelocytic Leukemia. Acta Haematol. 2013, 130, 260–267. [Google Scholar] [CrossRef]
  540. Chang, K.J.; Yin, J.Z.; Huang, H.; Li, B.; Yang, M.H. Arsenic Trioxide Inhibits the Growth of Cancer Stem Cells Derived from Small Cell Lung Cancer by Downregulating Stem Cell- Maintenance Factors and Inducing Apoptosis via the Hedgehog Signaling Blockade. Transl. Lung Cancer Res. 2020, 9, 1379–1396. [Google Scholar] [CrossRef]
  541. Beauchamp, E.M.; Ringer, L.; Bulut, G.; Sajwan, K.P.; Hall, M.D.; Lee, Y.C.; Peaceman, D.; Özdemirli, M.; Rodriguez, O.; Macdonald, T.J.; et al. Arsenic Trioxide Inhibits Human Cancer Cell Growth and Tumor Development in Mice by Blocking Hedgehog/GLI Pathway. J. Clin. Investig. 2011, 121, 148. [Google Scholar] [CrossRef] [PubMed]
  542. Bin Han, J.; Sang, F.; Chang, J.J.; Hua, Y.Q.; Shi, W.D.; Tang, L.H.; Liu, L. ming. Arsenic Trioxide Inhibits Viability of Pancreatic Cancer Stem Cells in Culture and in a Xenograft Model via Binding to SHH-Gli. Onco Targets Ther. 2013, 6, 1129. [Google Scholar] [CrossRef] [PubMed]
  543. Lauth, M.; Bergström, Å.; Shimokawa, T.; Toftgård, R. Inhibition of GLI-Mediated Transcription and Tumor Cell Growth by Small-Molecule Antagonists. Proc. Natl. Acad. Sci. USA 2007, 104, 8455–8460. [Google Scholar] [CrossRef] [PubMed]
  544. Kiesslich, T.; Mayr, C.; Wachter, J.; Bach, D.; Fuereder, J.; Wagner, A.; Alinger, B.; Pichler, M.; Di Fazio, P.; Ocker, M.; et al. Activated Hedgehog Pathway Is a Potential Target for Pharmacological Intervention in Biliary Tract Cancer. Mol. Cell Biochem. 2014, 396, 257–268. [Google Scholar] [CrossRef] [PubMed]
  545. Harada, K.; Ohashi, R.; Naito, K.; Kanki, K. Hedgehog Signal Inhibitor GANT61 Inhibits the Malignant Behavior of Undifferentiated Hepatocellular Carcinoma Cells by Targeting Non-Canonical GLI Signaling. Int. J. Mol. Sci. 2020, 21, 3126. [Google Scholar] [CrossRef] [PubMed]
  546. Michaud, N.R.; Wang, Y.; McEachern, K.A.; Jordan, J.J.; Mazzola, A.M.; Hernandez, A.; Jalla, S.; Chesebrough, J.W.; Hynes, M.J.; Belmonte, M.A.; et al. Novel Neutralizing Hedgehog Antibody Medi-5304 Exhibits Antitumor Activity by Inhibiting Paracrine Hedgehog Signaling. Mol. Cancer Ther. 2014, 13, 386–398. [Google Scholar] [CrossRef] [PubMed]
  547. Bausch, D.; Fritz, S.; Bolm, L.; Wellner, U.F.; Fernandez-del-Castillo, C.; Warshaw, A.L.; Thayer, S.P.; Liss, A.S. Hedgehog Signaling Promotes Angiogenesis Directly and Indirectly in Pancreatic Cancer. Angiogenesis 2020, 23, 479–492. [Google Scholar] [CrossRef] [PubMed]
  548. O’Toole, S.A.; Machalek, D.A.; Shearer, R.F.; Millar, E.K.A.; Nair, R.; Schofield, P.; McLeod, D.; Cooper, C.L.; McNeil, C.M.; McFarland, A.; et al. Hedgehog Overexpression Is Associated with Stromal Interactions and Predicts for Poor Outcome in Breast Cancer. Cancer Res. 2011, 71, 4002–4014. [Google Scholar] [CrossRef]
  549. Bissey, P.A.; Mathot, P.; Guix, C.; Jasmin, M.; Goddard, I.; Costechareyre, C.; Gadot, N.; Delcros, J.G.; Mali, S.M.; Fasan, R.; et al. Blocking SHH/Patched Interaction Triggers Tumor Growth Inhibition through Patched-Induced Apoptosis. Cancer Res. 2020, 80, 1970–1980. [Google Scholar] [CrossRef]
  550. Nakamura, M.; Kubo, M.; Yanai, K.; Mikami, Y.; Ikebe, M.; Nagai, S.; Yamaguchi, K.; Tanaka, M.; Katano, M. Anti-Patched-1 Antibodies Suppress Hedgehog Signaling Pathway and Pancreatic Cancer Proliferation. Anticancer Res. 2007, 27, 3743–3747. [Google Scholar] [PubMed]
Figure 1. Cellular localization of CSC markers, pathways, and factors associated with CSCs in solid tumors as therapeutic targets. Schematic representation depicting the intracellular and membrane-bound distribution of key CSC markers, pathways, and factors associated with CSCs ALDH1A1, ALDH1A3, ALDH2, ALDH3A1, EpCAM, CD44, CD133, CD55, CXCR4, SOX2, Notch, Nanog, and SHH. Arrows depict signaling pathway intermediates.
Figure 1. Cellular localization of CSC markers, pathways, and factors associated with CSCs in solid tumors as therapeutic targets. Schematic representation depicting the intracellular and membrane-bound distribution of key CSC markers, pathways, and factors associated with CSCs ALDH1A1, ALDH1A3, ALDH2, ALDH3A1, EpCAM, CD44, CD133, CD55, CXCR4, SOX2, Notch, Nanog, and SHH. Arrows depict signaling pathway intermediates.
Ijms 25 04102 g001
Figure 2. Notch signaling pathway and therapeutic interventions. Schematic representation depicting the therapeutic targets of the Notch signaling pathway. Delta-like ligand binding to the Notch receptor results in proteolytic cleavage of the Notch receptor, catalyzed by ADAM metalloproteases and γ-secretase to release the Notch intracellular domain. DLL: Delta-like ligand, NICD: Notch intracellular domain, ADAM17: A disintegrin and metalloproteinase 17, MAML: mastermind-like, CSL: CFB1/RBPJκ/Su(H)/LAG-1.
Figure 2. Notch signaling pathway and therapeutic interventions. Schematic representation depicting the therapeutic targets of the Notch signaling pathway. Delta-like ligand binding to the Notch receptor results in proteolytic cleavage of the Notch receptor, catalyzed by ADAM metalloproteases and γ-secretase to release the Notch intracellular domain. DLL: Delta-like ligand, NICD: Notch intracellular domain, ADAM17: A disintegrin and metalloproteinase 17, MAML: mastermind-like, CSL: CFB1/RBPJκ/Su(H)/LAG-1.
Ijms 25 04102 g002
Figure 3. Therapeutic targets of the Wnt/β-catenin signaling pathway. Wnt ligands are activated by porcupine-mediated lipid modification and bind to Frizzled receptors and LRP co-receptors. R-spondin binding to Lgr receptors enhances Wnt signaling by sequestering ZNRF3 and RNF43 to stabilize the Wnt/Fzd interaction. Activation of the Fzd/LRP receptor complex leads to the recruitment and activation of Disheveled. In turn, Disheveled inhibits the β-catenin destruction complex. β-catenin translocates to the nucleus, where it displaces TBL1-containing corepressor complexes from TCF/LEF transcription factors to allow the transcription of target genes. CBP is recruited to the promoter regions of Wnt target genes. PORCN: Porcupine, Wnt: wingless-related integration site, LRP: low-density lipoprotein receptor-related protein, ZNRF3: zinc and ring finger 3, RNF43: ring finger protein 43, Lgr: leucine-rich repeat-containing G protein-coupled receptor, RSPO: R-spondin, GSK-3β: glycogen synthase kinase-3β, Axin: axis inhibition protein, APC: adenomatous polyposis coli, CK1α: casein kinase 1 α, TBL1: transducin beta-like 1, TCF/LEF: T-cell factor/lymphoid enhancer factor, CBP: CREB-binding protein.
Figure 3. Therapeutic targets of the Wnt/β-catenin signaling pathway. Wnt ligands are activated by porcupine-mediated lipid modification and bind to Frizzled receptors and LRP co-receptors. R-spondin binding to Lgr receptors enhances Wnt signaling by sequestering ZNRF3 and RNF43 to stabilize the Wnt/Fzd interaction. Activation of the Fzd/LRP receptor complex leads to the recruitment and activation of Disheveled. In turn, Disheveled inhibits the β-catenin destruction complex. β-catenin translocates to the nucleus, where it displaces TBL1-containing corepressor complexes from TCF/LEF transcription factors to allow the transcription of target genes. CBP is recruited to the promoter regions of Wnt target genes. PORCN: Porcupine, Wnt: wingless-related integration site, LRP: low-density lipoprotein receptor-related protein, ZNRF3: zinc and ring finger 3, RNF43: ring finger protein 43, Lgr: leucine-rich repeat-containing G protein-coupled receptor, RSPO: R-spondin, GSK-3β: glycogen synthase kinase-3β, Axin: axis inhibition protein, APC: adenomatous polyposis coli, CK1α: casein kinase 1 α, TBL1: transducin beta-like 1, TCF/LEF: T-cell factor/lymphoid enhancer factor, CBP: CREB-binding protein.
Ijms 25 04102 g003
Figure 4. Therapeutic targets in the HH signaling pathway. Hedgehog ligands bind to Patched on the surface of target cells, relieving Patched-mediated inhibition of Smoothened. Activated Smoothened leads to activation of the Gli family of transcription factor, which promotes the expression of target genes. PTCH: Patched, SMO: smoothened, GLI1: Glioma-associated oncogene homolog 1.
Figure 4. Therapeutic targets in the HH signaling pathway. Hedgehog ligands bind to Patched on the surface of target cells, relieving Patched-mediated inhibition of Smoothened. Activated Smoothened leads to activation of the Gli family of transcription factor, which promotes the expression of target genes. PTCH: Patched, SMO: smoothened, GLI1: Glioma-associated oncogene homolog 1.
Ijms 25 04102 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

MacLean, M.R.; Walker, O.L.; Arun, R.P.; Fernando, W.; Marcato, P. Informed by Cancer Stem Cells of Solid Tumors: Advances in Treatments Targeting Tumor-Promoting Factors and Pathways. Int. J. Mol. Sci. 2024, 25, 4102. https://doi.org/10.3390/ijms25074102

AMA Style

MacLean MR, Walker OL, Arun RP, Fernando W, Marcato P. Informed by Cancer Stem Cells of Solid Tumors: Advances in Treatments Targeting Tumor-Promoting Factors and Pathways. International Journal of Molecular Sciences. 2024; 25(7):4102. https://doi.org/10.3390/ijms25074102

Chicago/Turabian Style

MacLean, Maya R., Olivia L. Walker, Raj Pranap Arun, Wasundara Fernando, and Paola Marcato. 2024. "Informed by Cancer Stem Cells of Solid Tumors: Advances in Treatments Targeting Tumor-Promoting Factors and Pathways" International Journal of Molecular Sciences 25, no. 7: 4102. https://doi.org/10.3390/ijms25074102

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