Open Access

Roles of DEPDC1 in various types of cancer (Review)

  • Authors:
    • Danqi Liu
    • Haima Li
    • Jia Ouyang
  • View Affiliations

  • Published online on: August 29, 2024     https://doi.org/10.3892/ol.2024.14651
  • Article Number: 518
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Dishevelled, EGL‑10 and pleckstrin domain-containing 1 (DEPDC1) has been identified as a crucial factor in the development and progression of various types of cancer. This protein, which is largely undetectable in normal tissues but is highly expressed in numerous tumor types, serves a significant role in cell mitosis, proliferation, migration, invasion, angiogenesis, autophagy and apoptosis. Furthermore, DEPDC1 is implicated in several key signaling pathways, such as NF‑κB, PI3K/Akt, Wnt/β‑catenin and Hippo pathways, which are essential for cell proliferation and survival. The expression of DEPDC1 has been linked to poor prognosis and survival rates in multiple types of cancer, including hepatocellular carcinoma, lung adenocarcinoma, colorectal cancer and breast cancer. Notably, DEPDC1 has been suggested to have potential as a diagnostic and prognostic marker, as well as a therapeutic target. Its involvement in critical signaling pathways suggests that targeting DEPDC1 could inhibit tumor growth and metastasis, thereby improving patient outcomes. In addition, clinical trials have shown promising results for DEPDC1‑derived peptide vaccines, indicating their safety and potential efficacy in cancer treatment. To the best of our knowledge, this is the first comprehensive review addressing the role of DEPDC1 in cancer. Through a critical analysis of existing studies, the present review aimed to consolidate existing knowledge and highlight gaps in understanding, paving the way for future research to elucidate the complex interactions of DEPDC1 in the context of cancer biology.

Introduction

The disheveled, EGL-10 and pleckstrin (DEP) globular protein domain interacts with phospholipids and membrane receptors to recruit proteins to the plasma membrane for participation in signal transduction (1). As first described by Kanehira et al (2) in 2007, knockdown of upregulated DEP domain-containing 1 (DEPDC1) was shown to significantly suppress the proliferation of bladder cancer (BC) cells. Subsequent studies (39) have implicated DEPDC1 in multiple signaling pathways associated with tumorigenesis.

DEPDC1, also named DEPDC1A, which is located on human chromosome 1p31.3 (2), and DEPDC1B, located on chromosome 5q12.1 (10), are responsible for cell cycle regulation (4,11). There are two isoforms of DEPDC1A: DEPDC1-V1 and DEPDC1-V2. DEPDC1-V1 is composed of 5,318 nucleotides that encode 811 amino acids comprising 12 exons, while DEPDC1-V2 is composed of 4,466 nucleotides that encode 527 amino acids compromising 11 exons (DEPDC1-V2 lacks exon 8 of DEPDC1-V1) (Fig. 1).

DEPDC1 is considered a cancer-testis antigen (3), which means that other than the testes, DEPDC1 is barely detectable in normal human tissues; however, it is highly expressed in a number of tumors (2). Notably, DEPDC1 is a highly conserved protein and expression of both its isoforms is increased during mitosis (12). DEPDC1 is highly expressed in the nucleus during prophase and redistributed throughout the cell after breakdown of the nuclear membrane in the middle and late stages of mitosis (13).

DEPDC1 serves critical roles in cell mitosis, proliferation, migration and invasion, as well as angiogenesis, autophagy and apoptosis (3,1215). Therefore, DEPDC1 may be considered a potential marker of prognosis and response to therapy. In the present review, the roles of DEPDC1 in tumorigenesis and its related molecular mechanisms, as well as its potential as a peptide vaccine are summarized to emphasize the importance of DEPDC1 in future cancer research.

Association of DEPDC1 with tumorigenesis

Upregulation of DEPDC1 has been associated with poor prognosis and survival in hepatocellular carcinoma (HCC) (1621), lung adenocarcinoma (LUAD) (22,23), colorectal cancer (CRC) (2426), breast cancer (BrC) (2730), ovarian cancer (31), renal cancer (32), osteosarcoma (33,34), anaplastic thyroid carcinoma (35), gastric cancer (36), cholangiocarcinoma (37), head and neck squamous cell carcinoma (38), esophageal squamous cell carcinoma (39), endometrial carcinoma (40), multiple myeloma (4,41), glioma (42), diffuse large B-cell lymphoma (43) and soft-tissue sarcoma (44). Furthermore, DEPDC1 has been reported to be significantly upregulated in 29 out of 33 types of human cancer, and to be associated with overall survival, disease-specific survival and progress-free interval prognosis in numerous tumor types (45). The upregulation of DEPDC1 may be related to apoptosis, autophagy, proliferation, migration, invasion and angiogenesis in various types of cancer (4649). Hence, the present study reviewed the molecular mechanisms underlying the multiple functions of DEPDC1 in tumorigenesis.

Molecular mechanisms of DEPDC1 in tumorigenesis

BC

BC is the 10th most common cancer and the second most common cancer in men worldwide (50). The role of DEPDC1 in BC was first described in 2007. Harada et al (3) reported that DEPDC1 forms a complex with zinc finger protein 224 (ZNF224) that suppresses transcription of A20, resulting in inhibition of apoptosis via activation of NF-κB and the subsequent proliferation of BC cells, whereas a peptide mimic formed by coupling 11 arginine residues at the amino terminal of DEPDC1 (nucleotides 611–628) was able to inhibit formation of the DEPDC1-ZNF224 complex and induce apoptosis of BC cells both in vivo and in vitro. Wang et al (51) revealed that overexpression of DEPDC1 could alleviate or even reverse the inhibition of proliferation and migration of BC cells induced by knockdown of α-protein kinase 2.

Lung cancer (LC)

Globally, LC remains a leading cause of cancer-related death in both men and women (52). The two main types of LC are small cell and non-small cell subtypes. Similar to BC, Wang et al (9) reported activation of the DEPDC1-ZNF224/A20/NF-κB axis in A549 LUAD cells. Maternal embryonic leucine zipper kinase (MELK) is a is a highly conserved serine/threonine kinase in various stages of the cell cycle (53,54). Knockout of MELK in A549 cells has been reported to significantly reduce DEPDC1 protein levels without any change to transcription levels (55). In addition, MELK-induced phosphorylation can regulate DEPDC1 protein stability (55), similar to the findings of a previous report in myeloma cells (56). Wang et al (15) reported that DEPDC1 could upregulate the expression of RAS and suppress autophagy via the RAS-ERK pathway in LUAD cells, thus revealing a potential relationship between DEPDC1 and autophagy.

MicroRNAs (miRNAs/miRs) are short non-coding RNAs that serve crucial roles in oncogenesis and have been investigated as potential diagnostic and prognostic markers. A study by Liu et al (57) found that miR-23b could downregulate DEPDC1 in non-small cell LC. Multiple miRNAs have been reported to inhibit the expression of DEPDC1 in various types of cancer, as described below.

HCC

HCC is one of the most common causes of cancer-related mortality worldwide, accounting for >800,000 deaths annually, with a 1-year survival rate of <20% (58). A study by Guo et al (14) revealed that upregulation of DEPDC1 induced the expression of phosphorylated (p)-Akt, c-myc and cyclin E1 in HCC cells, whereas knockdown of chemokine ligand 20 (CCL20) or chemokine receptor 6 (CCR6; receptor for CCL20) reversed these effects. In addition, knockdown of CCL20 or CCR6 reversed DEPDC1-related angiogenesis and the invasive capabilities of human umbilical vein endothelial cells. These results indicated that DEPDC1 may promote angiogenesis in HCC.

Li et al (59) and Qu et al (60) reported that DEPDC1 promoted the proliferation and metastasis of HCC via activation of the Wnt/β-catenin signaling pathway. Zhou et al (61) also demonstrated that DEPDC1 promoted the proliferation of HCC cells and suppressed chemotherapy sensitivity via activation of the JNK signaling pathway.

Two studies reported on the regulation of DEPDC1 by non-coding RNA in HCC. Tian et al (49) revealed that long intergenic non-protein coding RNA, regulator of reprogramming promoted the stability of DEPDC1 mRNA via heterogeneous nuclear ribonucleoprotein K and could act as a competing endogenous RNA that affects the function of DEPDC1. Xu et al (62) demonstrated that certain miRNAs (including miR-96, miR-145 and miR-183) and mRNAs (including NAT2, FBXO5, CCNB1, DEPDC1 and NTN4) may be associated with the efficacy of ribavirin for treatment of HCC; among them, miR-96 and DEPDC1 constituted a miRNA-mRNA regulation pair.

CRC

CRC is the second most common malignant tumor worldwide, the incidence rate of which has increased annually (63). Wang et al (48) reported that DEPDC1-induced enhanced expression of suppressor of zest 12 could promote the proliferation, invasion and epithelial-mesenchymal transition of CRC cells. Sharen et al (64) revealed that overexpression of the large ribosomal subunit protein eL31 promoted the proliferation and migration of CRC cells by targeting DEPDC1, and upregulated the expression of p-Akt, cyclin D1, cyclin-dependent kinase 6, phosphatidylinositol-4,5-bisphosphate 3-kinase and catalytic subunit α.

Zhao et al (65) discovered that sirtuin 1 can bind to the promoter of miR-20b-3p and inhibit the expression of miR-20b-3p. Notably, miR-20b-3p negatively regulates DEPDC1, and DEPDC1 upregulation can enhance the resistance of CRC to oxaliplatin. Lou et al (66) suggested that miR-455-5p enhanced the sensitivity of CRC cells to 5-fluorouracil via negative regulation of PI3K regulatory subunit α and DEPDC1 independently.

BrC

BrC has been reported to be the most commonly diagnosed type of cancer, accounting for 2.26 million cases worldwide in 2020, and is the leading cause of cancer-related death of women (67). Zhao et al (68) demonstrated that DEPDC1 promoted the proliferation, migration and invasion of BrC cells via the PI3K/Akt/mammalian target of rapamycin (mTOR) signaling pathway.

Zhang et al (69) reported that miR-26b suppressed the expression of DEPDC1 in triple-negative BrC (TNBC), and DEPDC1 promoted the growth and proliferation of TNBC cells by increasing the expression of forkhead box protein (FOX)M1. Hao et al (47) reported that miR-374c-5p inhibited the progression of BrC via TATA-box binding protein associated factor 7-mediated downregulation of DEPDC1.

Prostate cancer (PC)

PC is an important cause of male mortality worldwide (70). Huang et al (8) reported that DEPDC1 increased the activity of transcription factor E2F1, and subsequently upregulated the expression of cyclin D1 and cyclin-dependent kinase 2, leading to more frequent G1-S phase cell cycle transition in PC. Ramalho-Carvalho et al (71) demonstrated that miR-130a inhibited DEPDC1 expression in PC cells.

Oral squamous cell carcinoma (OSCC)

OSCC is a major public health concern, accounting for >90% of all oral malignancies worldwide (72). Guo et al (46) indicated that the tobacco-specific nitrosamine carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone could lead to DNA methyltransferase 1-regulated DNA methylation, which may promote DEPDC1 expression; this upregulated DEPDC1 expression can stimulate the proliferation of OSCC cells by inhibiting the expression of cytochrome P450 family 27 subfamily B member 1. Qiu et al (73) suggested that DEPDC1 may positively regulate the expression of FOXM1, resulting in nuclear localization of β-catenin, which is the most important indicator of Wnt pathway activation. Huang et al (74) revealed that DEPDC1 promoted activation of the Wnt/β-catenin signaling pathway in OSCC.

Nephroblastoma (NB)

NB, or Wilms tumor, is the most common renal cell malignancy and the second most common extracranial solid tumor in children after neuroblastoma, (75). Geng et al (76) demonstrated that, in NB cells, overexpression of FOXO3a inhibited the protein expression of p-GSK-3β, Wnt3a and β-catenin, whereas overexpression of DEPDC1 had the opposite effects. In addition, FOXO3a inhibited the transcription of DEPDC1, which was associated with NB-like proliferation, migration and invasion. This previous study also revealed that knockout of S100 calcium-binding protein A16 (S100A16) inhibited the migration, invasiveness and angiogenesis of NB cells, which was partially reversed by overexpression of DEPDC1. Furthermore, a study by Geng et al (77) demonstrated that S100A16 and DEPDC1 promoted the progression and angiogenesis of NB through the PI3K/Akt/mTOR pathway.

Other types of cancer

A study by Yang et al (7) showed that downregulation of DEPDC1 reversed the inhibitory effect of protocadherin-10 (PCDH10) against the proliferation of endometrial endometrioid carcinoma cells, and knockdown of DEPDC1 blocked progression of the cell cycle and promoted apoptosis, thus replicating the effects of PCDH10 overexpression and indicating that PCDH10 may be upstream of DEPDC1.

Feng et al (78) demonstrated that knockout of DEPDC1 inhibited the cell cycle progression of nasopharyngeal carcinoma and the occurrence of nasopharyngeal carcinoma tumor formation in nude mice. However, the expression levels of cell cycle-related genes were measured in nasopharyngeal carcinoma cells after knockout of DEPDC1, without exploring the specific molecular mechanisms. Meanwhile, Kikuchi et al (79) revealed that DEPDC1 inhibited the apoptosis of glioma cells via the NF-κB signaling pathway.

Yang et al (80) demonstrated that downregulation of DEPDC1 activated the Hippo signaling pathway by overexpressing p-large tumor suppressor kinase 1 and p-yes-associated protein 1, thereby inhibiting the proliferation, invasion, migration and angiogenesis of osteosarcoma cells. Furthermore, overexpression of kinesin family member 4A reversed the effects of DEPDC1 on downregulation of the Hippo signaling pathway.

These studies have indicated that DEPDC1 serves a complex and critical role in several important signaling pathways in various types of cancer (Fig. 2; Table I); however, the molecular mechanisms require further clarification.

Table I.

Molecular mechanism of DEPDC1 in various tumors.

Table I.

Molecular mechanism of DEPDC1 in various tumors.

Cancer typePossible mechanisms(Refs.)
BCDEPDC1-ZNF224 complex inhibits A20 transcription, thereby activating the NF-κB pathway to inhibit apoptosis; 11R-DEP: 611–628 peptide induces apoptosis by inhibiting the formation of the aforementioned complex(3)
ALPK2 promotes proliferation and migration by positively regulating DEPDC1(51)
Lung cancerDEPDC1-ZNF224 complex inhibits A20 transcription, thereby activating the NF-κB pathway to inhibit apoptosis; 11R-DEP: 611–628 peptide induces apoptosis by inhibiting the formation of the aforementioned complex(9)
MELK enhances DEPDC1 protein stability(55)
DEPDC1 suppresses autophagy via the RAS-ERK pathway(15)
STAT1 induces upregulation of KTN1-AS1, which inhibits the expression of miR-23b and thus upregulates DEPDC1(57)
HCCDEPDC1 upregulates CCL20/CCR6 to promote angiogenesis(14)
DEPDC1 activates the Wnt/β-catenin pathway(59,60)
DEPDC1 activates the JNK pathway(61)
Linc-ROR upregulates DEPDC1 by competitively binding to miR-130a-3p, inducing EMT and angiogenesis(49)
miR-96 and DEPDC1 constitute a miRNA-mRNA regulation pair(62)
CRCDEPDC1 upregulates SUZ12 and H3K27Me3 to promote EMT(48)
eL31 promotes proliferation and migration by targeting DEPDC1(64)
SIRT1 mediates DEPDC1 upregulation by inhibiting the expression of miR-20b-3p(65)
miR-455-5p inhibits DEPDC1 expression(66)
Breast cancerDEPDC1 activates the PI3K/AKT/mTOR pathway(68)
miR-26b negatively regulates the expression of DEPDC1, and DEPDC1 positively regulates the expression of FOXM1(69)
miR-374c-5p downregulates DEPDC1 by inhibiting TAF7 expression(47)
PCDEPDC1 interacts with E2F1 and increases its transcriptional activity to upregulate cyclin D1 and CDK2 to promote the cell cycle(8)
DEPDC1 is suppressed by miR-130a(71)
OSCCNNK promotes the expression of DEPDC1 through DNMT1, thereby inhibiting the expression of CYP27B1(46)
DEPDC1 activates the Wnt/β-catenin pathway through FOXM1(73)
DEPDC1 activates the Wnt/β-catenin pathway(74)
NBFOXO3a inhibits the Wnt/β-catenin pathway by downregulating DEPDC1(76)
S100A16 acts upstream of DEPDC1 and promotes angiogenesis through the PI3K/Akt/mTOR pathway(77)
EECPCDH10-induced downregulation of DEPDC1 induces apoptosis(7)
GliomaDEPDC1 activates the NF-κB signaling pathway by inhibiting A20(79)
OsteosarcomaDEPDC1 inhibits the Hippo signaling pathway by upregulating KIF4A(80)

[i] DEPDC1, dishevelled, EGL-10 and pleckstrin domain-containing 1; EMT, epithelial-mesenchymal transition; BC, bladder cancer; HCC, hepatocellular carcinoma; CRC, colorectal cancer; PC, prostate cancer; OSCC, oral squamous cell carcinoma; NB, nephroblastoma; EEC, endometrial endometrioid carcinoma.

DEPDC1 antisense long non-coding (lnc)RNA (DEPDC1-AS1)

LncRNAs are involved in a variety of cell signaling pathways. Some lncRNAs act as carcinogens in tumors, whereas others act as tumor suppressors (81). Antisense lncRNAs are transcribed from the DNA lagging strand as complementary sequences to the leading strand (82).

DEPDC1-AS1 is considered a reliable prognostic marker of LUAD (83) and ovarian cancer (84). DEPDC1-AS1 has been reported to be associated with ferroptosis and to have the highest hazard ratio among the 12 best prognostic markers of LUAD (85). Xu et al (86) reported that DEPDC1-AS1 may be considered a reliable marker for the detection and prognosis of gastric cancer, where it can bind to human antigen and increase the stability of F11 receptor mRNA, which is associated with the proliferation and metastasis of gastric cancer cells.

Relatively few studies (8386) have investigated the role of DEPDC1-AS1 in tumorigenesis; therefore, the specific molecular mechanisms remain unclear. It has been suggested that antisense lncRNAs may act through RNA-DNA hybridization (87). Notably, the effect of DEPDC1-AS1 on DEPDC1 expression remains unknown; however, DEPDC1-AS1 has potential for the detection and prognosis of various types of cancer, although further studies are needed to elucidate the underlying mechanisms.

Clinical significance of DEPDC1

DEPDC1 as a diagnostic and prognostic marker

As aforementioned, high DEPDC1 expression is associated with the poor prognosis of various types of cancer. Therefore, DEPDC1 may have potential as a diagnostic and prognostic marker for clinical diagnosis and response to treatment. For example, a significant proportion of LUAD cases are caused by mutations to epidermal growth factor receptor, K-Ras or anaplastic lymphoma kinase; however, these three genes are not suitable as markers of triple-negative LUAD. Alternatively, high expression of DEPDC1 has been associated with the poor prognosis of triple-negative LUAD (5) and may thus be a promising target for cancer prognosis.

Potential therapeutic targets

DEPDC1 is related to multiple signaling pathways and may be a signaling hub for tumor development, and could thus be considered a promising new therapeutic target. Arumugam et al (88) suggested that DEPDC1, as a consistently differentially expressed gene in TNBC, may interact well with doxorubicin and anethole, which is a phytocompound, suggesting that DEPDC1 could be a possible therapeutic target of TNBC.

DEPDC1 is also a potential marker of the response to medications. Microtubule-targeted chemotherapy induces apoptosis of cancer cells by promoting phosphorylation and degradation of induced myeloid leukemia cell differentiation protein Mcl (MCL1), a member of the anti-apoptotic Bcl-2 family (89). Sendoel et al (6) concluded that DEPDC1 may regulate vincristine (a tubulin-targeted chemotherapy drug)-induced cell death by promoting JNK-dependent degradation of MCL1, whereas knockdown of DEPDC1 mediated resistance to vincristine-induced cell death, suggesting that vincristine may serve an antitumor role through DEPDC1. Therefore, whether the expression levels of DEPDC1 can predict the efficacy of anti-tubulin drugs on tumors may be a meaningful research direction.

As aforementioned, multiple miRNAs can inhibit the expression of DEPDC1 and enhance the sensitivity of tumors to chemotherapy drugs to address drug resistance. Therefore, the use of miRNAs targeting DEPDC1 may be a promising research direction to reduce chemotherapy resistance.

Notably, a variety of Chinese herbal extracts, such as paeoniflorin (90), a compound found in Paeonia lactiflora and Paeonia × suffruticosa, and platycodin D (91), a bioactive component of the roots of Platycodon grandifloras, have been reported to exhibit anticancer effects via multiple signaling pathways. Therefore, it may be possible to explore whether there are natural drugs that can inhibit DEPDC1. Therefore, future studies are warranted to verify the feasibility of DEPDC1-targeted anticancer therapies.

Peptide vaccines

Peptide-based anticancer vaccines use tumor-associated or tumor-specific peptides to induce and activate T cells. These peptides are present on human leukocyte antigen (HLA) molecules on the cell surface and are recognized by T-cell receptors. Peptide vaccines have the advantages of low side effects, but must be optimized prior to clinical application (92).

Shortly after being described in BC, DEPDC1 was investigated as a new target for peptide vaccines. Obara et al (93) conducted a clinical trial of possible peptide fragments that would bind to the HLA-A*2402 molecule, and administered the M phase phosphoprotein 1 (MPHOSPH1)-278 and DEPDC1-294 peptides to six patients with BC. Four of the six patients vaccinated with DEPDC1-294 achieved cytotoxic T lymphocyte (CTL)-positive responses to DEPDC1-294, with one achieving a partial response. This sample was expanded in a further study to 32 patients who received this combined peptide vaccine, and the study concluded that the 2-year survival rate was slightly better than in response to treatment with vinflunine (94). However, this previous study did not include a control group, but rather compared the efficacy of vinflunine with other studies, which may lead to errors due to different backgrounds of the patients (94). In addition, the ability of the peptide vaccine to prevent recurrence of non-muscular-invasive BC was evaluated after transurethral resection of bladder tumors in 127 patients (95). Combined with intravesical Bacillus Calmette-Guerin (BCG), the overall 2-year recurrence-free survival rate was 74% for patients in the HLA-A*2402-positive group, which was higher than the negative control group (95). However, both groups received the BCG vaccine, which might have masked the therapeutic effect of the peptide vaccine. Therefore, an additional study with three groups (peptide alone, BCG alone and peptide combined with BCG vaccine) is required.

Fujiwara et al (96) conducted a clinical trial of 35 patients with gastric cancer treated with a HLA-A24-binding peptide vaccine, which comprised a mixture of multiple peptides derived from DEPDC1, upregulated LC 10 (URLC10) epitope peptide, FOXM1, kinesin family member 20A (KIF20A) and vascular endothelial growth factor receptor-1 (VEGFR1). The study found that this peptide vaccine treatment was safe and promising to induce a specific T-cell response in patients with advanced gastric cancer. The study further demonstrated that the peptide vaccine was well tolerated in combination with S-1; however, this study, as an early exploratory study, did not show a clear clinical benefit of this combination therapy over S-1 alone due to sample size limitations (97). Although no notable antitumor effect or survival benefit was observed due to the small sample size and short duration, these trials demonstrated that the vaccine was safe with no serious adverse reactions, except for minor skin reactions, and patients with injection site reactions tended to achieve better outcomes. All of the tested peptide vaccines induced a specific T-cell response, and the patients who responded to CTL achieved better survival than those who did not, especially those who responded to the DEPDC1 peptide.

A clinical trial conducted by Daiko et al (98) of 13 patients with esophageal cancer treated with five peptides revealed that at least one peptide induced a response to CTL in all patients, although the CTL initiation rate of the DEPDC1 peptide was only 26.7%, and the efficacy was not analyzed, as only safety was addressed.

There have also been more successful treatments. For example, a 2016 study conducted by Murahashi et al (99) enlisted 18 patients treated with five HLA-A*2402 restricted tumor-associated antigen epitope peptides from the novel chloroplast outer membrane kinase KOC1, threonine tyrosine kinase, URLC10, DEPDC1 and MPHOSPH1 vaccines. A total of 4 days before vaccination, the patients received increasing dosages of cyclophosphamide. In this study, the overall survival of nine patients with CRC was 9.4 months, which was better than the effect of cetuximab in another study (6.1 months) (100), indicating the advantage of peptide vaccines with DEPDC1 as one of the epitope peptides. In addition, a 40-year-old male patient with esophageal cancer (celiac lymph node metastasis) achieved a complete response with no disease recurrence after 5 years.

In a 2019 study conducted by Kikuchi et al (101), 19 patients with grade III or IV glioma were treated with a peptide vaccine that included peptide epitopes from four glioma oncoantigens (lymphocyte antigen 6 family member K, DEPDC1, KIF20A and FOXM1) and two glioma angiogenesis-associated antigens (VEGFR1 and VEGFR2). Not only were there no serious adverse reactions, but a 33-year-old woman with anaplastic oligoastrocytoma (grade 3) was cured; their tumor shrank 3 months after vaccination and disappeared 9 months later with no tumor recurrence after 38 months. Although the sample size was small and there was no significant increase in overall survival, the fact that a patient was cured is encouraging, and further emphasizes the potential of DEPDC1 as a peptide vaccine target.

Some in vitro studies have reported that DEPDC1-derived peptides can induce activation of CTL and T helper (Th) cells (102,103). Furthermore, Yatsuda et al (104) demonstrated that the DEPDC1191-213 peptide induced a strong Th cell response in HLA-DR4 transgenic mice. As compared with CTL epitopes alone, co-inoculation of CTL and Th cell epitopes has also been reported to achieve clinical benefits in patients with melanoma (105). Therefore, co-vaccination with multiple CTL and Th epitope peptides containing DEPDC1 may induce a better immune response and result in better efficacy against tumors, although further clinical studies are needed.

Conclusion and future prospects

DEPDC1 is a newly discovered tumor-related gene that is upregulated in various malignant tumors and may be involved in tumorigenesis as a marker of poor prognosis. Therefore, the potential of DEPDC1 for detection and prognosis of cancer should be considered.

DEPDC1 is involved in the regulation of tumor cell proliferation, metastasis, autophagy, apoptosis and angiogenesis by mediating different signaling pathways. Some of these signaling pathways (NF-κB, PI3K/Akt, Wnt/β-catenin and Hippo) may promote cell proliferation and survival. However, since intracellular signal transduction is complex and there is cross-dialogue between different pathways, further studies are needed to clarify the signal transduction mechanism of DEPDC1.

Future studies may also focus on: i) Comparing the sensitivity and specificity of DEPDC1 with other common diagnostic and prognostic markers in different types of tumors; ii) investigating the unique role of DEPDC1 in predicting therapeutic response; and iii) evaluating the practical effectiveness of DEPDC1 as a marker in clinical applications.

Owing to the multifaceted characteristics and effects of cancer-testis antigens (3,102), DEPDC1 is a potential therapeutic target. Treatment targeting DEPDC1 could inhibit the proliferation and metastasis of tumor cells and improve patient outcomes. Furthermore, the efficacy of DEPDC1-derived peptides in immunotherapy as cancer vaccines has been confirmed in clinical trials, thus warranting further studies to identify effective peptide segments, determine appropriate dosages and assess efficacy in combination with other peptide vaccines, as well as radiotherapy and chemotherapy. DEPDC1 also provides a direction for the study of the molecular mechanisms of antitumor drugs. Clarification of the mechanism of DEPDC1 could provide a new therapeutic target for the prevention and treatment of malignant tumors.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 82104603) and the Peking University People's Hospital Research and Development Fund (grant nos. RS2021-12 and RDX2021-08).

Availability of data and materials

Not applicable.

Authors' contributions

DL performed the literature review and wrote the draft. HL was responsible for reviewing the literature, and reviewed and revised the draft. JO conceived and designed the study, acquired funding, and reviewed and revised the draft. Data authentication is not applicable. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Consonni SV, Maurice MM and Bos JL: DEP domains: Structurally similar but functionally different. Nat Rev Mol Cell Biol. 15:357–362. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Kanehira M, Harada Y, Takata R, Shuin T, Miki T, Fujioka T, Nakamura Y and Katagiri T: Involvement of upregulation of DEPDC1 (DEP domain containing 1) in bladder carcinogenesis. Oncogene. 26:6448–6455. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Harada Y, Kanehira M, Fujisawa Y, Takata R, Shuin T, Miki T, Fujioka T, Nakamura Y and Katagiri T: Cell-permeable peptide DEPDC1-ZNF224 interferes with transcriptional repression and oncogenicity in bladder cancer cells. Cancer Res. 70:5829–5839. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Kassambara A, Schoenhals M, Moreaux J, Veyrune JL, Rème T, Goldschmidt H, Hose D and Klein B: Inhibition of DEPDC1A, a bad prognostic marker in multiple myeloma, delays growth and induces mature plasma cell markers in malignant plasma cells. PLoS One. 8:e627522013. View Article : Google Scholar : PubMed/NCBI

5 

Okayama H, Kohno T, Ishii Y, Shimada Y, Shiraishi K, Iwakawa R, Furuta K, Tsuta K, Shibata T, Yamamoto S, et al: Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas. Cancer Res. 72:100–111. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Sendoel A, Maida S, Zheng X, Teo Y, Stergiou L, Rossi CA, Subasic D, Pinto SM, Kinchen JM, Shi M, et al: DEPDC1/LET-99 participates in an evolutionarily conserved pathway for anti-tubulin drug-induced apoptosis. Nat Cell Biol. 16:812–820. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Yang Y, Jiang Y, Jiang M, Zhang J, Yang B, She Y, Wang W, Deng Y and Ye Y: Protocadherin 10 inhibits cell proliferation and induces apoptosis via regulation of DEP domain containing 1 in endometrial endometrioid carcinoma. Exp Mol Pathol. 100:344–352. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Huang L, Chen K, Cai ZP, Chen FC, Shen HY, Zhao WH, Yang SJ, Chen XB, Tang GX and Lin X: DEPDC1 promotes cell proliferation and tumor growth via activation of E2F signaling in prostate cancer. Biochem Biophys Res Commun. 490:707–712. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Wang Q, Li A, Jin J and Huang G: Targeted interfering DEP domain containing 1 protein induces apoptosis in A549 lung adenocarcinoma cells through the NF-κB signaling pathway. Onco Targets Ther. 10:4443–4454. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Johannsdottir HK, Jonsson G, Johannesdottir G, Agnarsson BA, Eerola H, Arason A, Heikkila P, Egilsson V, Olsson H, Johannsson OT, et al: Chromosome 5 imbalance mapping in breast tumors from BRCA1 and BRCA2 mutation carriers and sporadic breast tumors. Int J Cancer. 119:1052–1060. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Zuo X, Wang D, Tao C, Dou X, Zhao Z, Zhang J, Huang S, Li Y, Zhang X, Bu Y and Wang Y: DEPDC1B is a novel direct target of B-Myb and contributes to malignant progression and immune infiltration in lung adenocarcinoma. Front Biosci (Landmark Ed). 29:2042024. View Article : Google Scholar : PubMed/NCBI

12 

Chen D, Ito S, Hyodo T, Asano-Inami E, Yuan H and Senga T: Phosphorylation of DEPDC1 at Ser110 is required to maintain centrosome organization during mitosis. Exp Cell Res. 358:101–110. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Mi Y, Zhang C, Bu Y, Zhang Y, He L, Li H, Zhu H, Li Y, Lei Y and Zhu J: DEPDC1 is a novel cell cycle related gene that regulates mitotic progression. BMB Rep. 48:413–418. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Guo W, Li H, Liu H, Ma X, Yang S and Wang Z: DEPDC1 drives hepatocellular carcinoma cell proliferation, invasion and angiogenesis by regulating the CCL20/CCR6 signaling pathway. Oncol Rep. 42:1075–1089. 2019.PubMed/NCBI

15 

Wang W, Li A, Han X, Wang Q, Guo J, Wu Y, Wang C and Huang G: DEPDC1 up-regulates RAS expression to inhibit autophagy in lung adenocarcinoma cells. J Cell Mol Med. 24:13303–13313. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Amisaki M, Yagyu T, Uchinaka EI, Morimoto M, Hanaki T, Watanabe J, Tokuyasu N, Sakamoto T, Honjo S and Fujiwara Y: Prognostic value of DEPDC1 expression in tumor and non-tumor tissue of patients with hepatocellular carcinoma. Anticancer Res. 39:4423–4430. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Yuan SG, Liao WJ, Yang JJ, Huang GJ and Huang ZQ: DEP domain containing 1 is a novel diagnostic marker and prognostic predictor for hepatocellular carcinoma. Asian Pac J Cancer Prev. 15:10917–10922. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Bin X, Luo Z, Wang J and Zhou S: Identification of a five immune term signature for prognosis and therapy options (immunotherapy versus targeted therapy) for patients with hepatocellular carcinoma. Comput Math Methods Med. 2023:89589622023. View Article : Google Scholar : PubMed/NCBI

19 

Zhang L, Li Y, Dai Y, Wang D, Wang X, Cao Y, Liu W and Tao Z: Glycolysis-related gene expression profiling serves as a novel prognosis risk predictor for human hepatocellular carcinoma. Sci Rep. 11:188752021. View Article : Google Scholar : PubMed/NCBI

20 

Pu Z, Zhu Y, Wang X, Zhong Y, Peng F and Zhang Y: Identification of prognostic biomarkers and correlation with immune infiltrates in hepatocellular carcinoma based on a competing endogenous RNA network. Front Genet. 12:5916232021. View Article : Google Scholar : PubMed/NCBI

21 

Zhang J, Liu X, Zhou W, Lu S, Wu C, Wu Z, Liu R, Li X, Wu J, Liu Y, et al: Identification of key genes associated with the process of hepatitis B inflammation and cancer transformation by integrated bioinformatics analysis. Front Genet. 12:6545172021. View Article : Google Scholar : PubMed/NCBI

22 

Shao F, Ling L, Li C, Huang X, Ye Y, Zhang M, Huang K, Pan J, Chen J and Wang Y: Establishing a metastasis-related diagnosis and prognosis model for lung adenocarcinoma through CRISPR library and TCGA database. J Cancer Res Clin Oncol. 149:885–899. 2023. View Article : Google Scholar : PubMed/NCBI

23 

Shen J and Xi M: DEPDC1 is highly expressed in lung adenocarcinoma and promotes tumor cell proliferation. Zhongguo Fei Ai Za Zhi. 24:453–460. 2021.(In Chinese). PubMed/NCBI

24 

Zhu Y, Sun L, Yu J, Xiang Y, Shen M, Wasan HS, Ruan S and Qiu S: Identification of biomarkers in colon cancer based on bioinformatic analysis. Transl Cancer Res. 9:4879–4895. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Shen X and Han J: Overexpression of gene DEP domain containing 1 and its clinical prognostic significance in colorectal cancer. J Clin Lab Anal. 34:e236342020. View Article : Google Scholar : PubMed/NCBI

26 

Miyata Y, Kumagai K, Nagaoka T, Kitaura K, Kaneda G, Kanazawa H, Suzuki S, Hamada Y and Suzuki R: Clinicopathological significance and prognostic value of Wilms' tumor gene expression in colorectal cancer. Cancer Biomark. 15:789–797. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Colak D, Nofal A, Albakheet A, Nirmal M, Jeprel H, Eldali A, Al-Tweigeri T, Tulbah A, Ajarim D, Malik OA, et al: Age-specific gene expression signatures for breast tumors and cross-species conserved potential cancer progression markers in young women. PLoS One. 8:e632042013. View Article : Google Scholar : PubMed/NCBI

28 

Kretschmer C, Sterner-Kock A, Siedentopf F, Schoenegg W, Schlag PM and Kemmner W: Identification of early molecular markers for breast cancer. Mol Cancer. 10:152011. View Article : Google Scholar : PubMed/NCBI

29 

Sang M, Wu M, Meng L, Zheng Y, Gu L, Liu F and Sang M: Identification of epithelial-mesenchymal transition-related circRNA-miRNA-mRNA ceRNA regulatory network in breast cancer. Pathol Res Pract. 216:1530882020. View Article : Google Scholar : PubMed/NCBI

30 

Kim J: In silico analysis of differentially expressed genesets in metastatic breast cancer identifies potential prognostic biomarkers. World J Surg Oncol. 19:1882021. View Article : Google Scholar : PubMed/NCBI

31 

Ke Y, Zhuang X and You L: Identification of core genes shared by endometrial cancer and ovarian cancer using an integrated approach. Cell Mol Biol (Noisy-le-grand). 68:140–145. 2022. View Article : Google Scholar : PubMed/NCBI

32 

Xu F, Guan Y, Xue L, Huang S, Gao K, Yang Z and Chong T: The effect of a novel glycolysis-related gene signature on progression, prognosis and immune microenvironment of renal cell carcinoma. BMC Cancer. 20:12072020. View Article : Google Scholar : PubMed/NCBI

33 

Ma Y, Guo J, Li D and Cai X: Identification of potential key genes and functional role of CENPF in osteosarcoma using bioinformatics and experimental analysis. Exp Ther Med. 23:802022. View Article : Google Scholar : PubMed/NCBI

34 

Shen L, Li H, Liu R, Zhou C, Bretches M, Gong X, Lu L, Zhang Y, Zhao K, Ning B, et al: DEPDC1 as a crucial factor in the progression of human osteosarcoma. Cancer Med. 12:5798–5808. 2023. View Article : Google Scholar : PubMed/NCBI

35 

Zhang Z, Zou Z, Dai H, Ye R, Di X, Li R, Ha Y, Sun Y and Gan S: Key genes involved in cell cycle arrest and DNA damage repair identified in anaplastic thyroid carcinoma using integrated bioinformatics analysis. Transl Cancer Res. 9:4188–4203. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Gong Z, Chu H, Chen J, Jiang L, Gong B, Zhu P, Zhang C, Wang Z, Zhang W, Wang J, et al: DEPDC1 upregulation promotes cell proliferation and predicts poor prognosis in patients with gastric cancer. Cancer Biomark. 30:299–307. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Mishra NK, Niu M, Southekal S, Bajpai P, Elkholy A, Manne U and Guda C: Identification of prognostic markers in cholangiocarcinoma using altered DNA methylation and gene expression profiles. Front Genet. 11:5221252020. View Article : Google Scholar : PubMed/NCBI

38 

Zhang J, Lin H, Jiang H, Jiang H, Xie T, Wang B, Huang X, Lin J, Xu A, Li R, et al: A key genomic signature associated with lymphovascular invasion in head and neck squamous cell carcinoma. BMC Cancer. 20:2662020. View Article : Google Scholar : PubMed/NCBI

39 

Zheng L, Li L, Xie J, Jin H and Zhu N: Six novel biomarkers for diagnosis and prognosis of esophageal squamous cell carcinoma: Validated by scRNA-seq and qPCR. J Cancer. 12:899–911. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Liu J, Wu Z, Sun R, Nie S, Meng H, Zhong Y, Nie X and Cheng W: Using mRNAsi to identify prognostic-related genes in endometrial carcinoma based on WGCNA. Life Sci. 258:1182312020. View Article : Google Scholar : PubMed/NCBI

41 

Noll JE, Vandyke K, Hewett DR, Mrozik KM, Bala RJ, Williams SA, Kok CH and Zannettino AC: PTTG1 expression is associated with hyperproliferative disease and poor prognosis in multiple myeloma. J Hematol Oncol. 8:1062015. View Article : Google Scholar : PubMed/NCBI

42 

Stangeland B, Mughal AA, Grieg Z, Sandberg CJ, Joel M, Nygård S, Meling T, Murrell W, Vik Mo EO and Langmoen IA: Combined expressional analysis, bioinformatics and targeted proteomics identify new potential therapeutic targets in glioblastoma stem cells. Oncotarget. 6:26192–26215. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Zhu QY: Bioinformatics analysis of the pathogenic link between Epstein-Barr virus infection, systemic lupus erythematosus and diffuse large B cell lymphoma. Sci Rep. 13:63102023. View Article : Google Scholar : PubMed/NCBI

44 

Pollino S, Benassi MS, Pazzaglia L, Conti A, Bertani N, Righi A, Piccinni-Leopardi M, Picci P and Perris R: Prognostic role of XTP1/DEPDC1B and SDP35/DEPDC1A in high grade soft-tissue sarcomas. Histol Histopathol. 33:597–608. 2018.PubMed/NCBI

45 

Jia B, Liu J, Hu X, Xia L and Han Y: Pan-cancer analysis of DEPDC1 as a candidate prognostic biomarker and associated with immune infiltration. Ann Transl Med. 10:13552022. View Article : Google Scholar : PubMed/NCBI

46 

Guo J, Zhou S, Huang P, Xu S, Zhang G, He H, Zeng Y, Xu CX, Kim H and Tan Y: NNK-mediated upregulation of DEPDC1 stimulates the progression of oral squamous cell carcinoma by inhibiting CYP27B1 expression. Am J Cancer Res. 10:1745–1760. 2020.PubMed/NCBI

47 

Hao S, Tian W, Chen Y, Wang L, Jiang Y, Gao B and Luo D: MicroRNA-374c-5p inhibits the development of breast cancer through TATA-box binding protein associated factor 7-mediated transcriptional regulation of DEP domain containing 1. J Cell Biochem. 120:15360–15368. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Wang Q, Jiang S, Liu J, Ma G, Zheng J and Zhang Y: DEP domain containing 1 promotes proliferation, invasion, and epithelial-mesenchymal transition in colorectal cancer by enhancing expression of suppressor of zest 12. Cancer Biother Radiopharm. 36:36–44. 2021.PubMed/NCBI

49 

Tian C, Abudoureyimu M, Lin X, Chu X and Wang R: Linc-ROR facilitates progression and angiogenesis of hepatocellular carcinoma by modulating DEPDC1 expression. Cell Death Dis. 12:10472021. View Article : Google Scholar : PubMed/NCBI

50 

Wang L, Wang Y, Wang J, Li L and Bi J: Identification of a prognosis-related risk signature for bladder cancer to predict survival and immune landscapes. J Immunol Res. 2021:32363842021. View Article : Google Scholar : PubMed/NCBI

51 

Wang Y, Wu J, Luo W, Zhang H, Shi G, Shen Y and Zhu Y, Ma C, Dai B, Ye D and Zhu Y: ALPK2 acts as tumor promotor in development of bladder cancer through targeting DEPDC1A. Cell Death Dis. 12:6612021. View Article : Google Scholar : PubMed/NCBI

52 

Barta JA, Powell CA and Wisnivesky JP: Global epidemiology of lung cancer. Ann Glob Health. 85:82019. View Article : Google Scholar : PubMed/NCBI

53 

Nakano I, Paucar AA, Bajpai R, Dougherty JD, Zewail A, Kelly TK, Kim KJ, Ou J, Groszer M, Imura T, et al: Maternal embryonic leucine zipper kinase (MELK) regulates multipotent neural progenitor proliferation. J Cell Biol. 170:413–427. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Ganguly R, Hong CS, Smith LGF, Kornblum HI and Nakano I: Maternal embryonic leucine zipper kinase: Key kinase for stem cell phenotype in glioma and other cancers. Mol Cancer Ther. 13:1393–1398. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Chung S, Kijima K, Kudo A, Fujisawa Y, Harada Y, Taira A, Takamatsu N, Miyamoto T, Matsuo Y and Nakamura Y: Preclinical evaluation of biomarkers associated with antitumor activity of MELK inhibitor. Oncotarget. 7:18171–18182. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Bolomsky A, Heusschen R, Schlangen K, Stangelberger K, Muller J, Schreiner W, Zojer N, Caers J and Ludwig H: Maternal embryonic leucine zipper kinase is a novel target for proliferation-associated high-risk myeloma. Haematologica. 103:325–335. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Liu C, Li X, Hao Y, Wang F, Cheng Z, Geng H and Geng D: STAT1-induced upregulation of lncRNA KTN1-AS1 predicts poor prognosis and facilitates non-small cell lung cancer progression via miR-23b/DEPDC1 axis. Aging (Albany NY). 12:8680–8701. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Johnson P, Zhou Q, Dao DY and Lo YMD: Circulating biomarkers in the diagnosis and management of hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 19:670–681. 2022. View Article : Google Scholar : PubMed/NCBI

59 

Li Y, Tian Y, Zhong W, Wang N, Wang Y, Zhang Y, Zhang Z, Li J, Ma F, Zhao Z and Peng Y: Artemisia argyi essential oil inhibits hepatocellular carcinoma metastasis via suppression of DEPDC1 dependent Wnt/β-catenin signaling pathway. Front Cell Dev Biol. 9:6647912021. View Article : Google Scholar : PubMed/NCBI

60 

Qu D, Cui F, Lu D, Yang Y and Xu Y: DEP domain containing 1 predicts prognosis of hepatocellular carcinoma patients and regulates tumor proliferation and metastasis. Cancer Sci. 110:157–165. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Zhou C, Wang P, Tu M, Huang Y, Xiong F and Wu Y: DEPDC1 promotes cell proliferation and suppresses sensitivity to chemotherapy in human hepatocellular carcinoma. Biosci Rep. 39:BSR201909462019. View Article : Google Scholar : PubMed/NCBI

62 

Xu C, Luo L, Yu Y, Zhang Z, Zhang Y, Li H, Cheng Y, Qin H, Zhang X, Ma H and Li Y: Screening therapeutic targets of ribavirin in hepatocellular carcinoma. Oncol Lett. 15:9625–9632. 2018.PubMed/NCBI

63 

Lewandowski M, Lipiński P, Bednarski I, Mik M and Dziki A: Knowledge and awareness of colorectal cancer. Pol Przegl Chir. 92:34–41. 2020. View Article : Google Scholar : PubMed/NCBI

64 

Sharen G, Li X, Sun J, Zhang L, Xi W, Zhao X, Han F, Jia L, A R, Cheng H and Hou M: Silencing eL31 suppresses the progression of colorectal cancer via targeting DEPDC1. J Transl Med. 20:4932022. View Article : Google Scholar : PubMed/NCBI

65 

Zhao B, Wang Y, Zhao X, Ni J, Zhu X, Fu Y and Yang F: SIRT1 enhances oxaliplatin resistance in colorectal cancer through microRNA-20b-3p/DEPDC1 axis. Cell Biol Int. 46:2107–2117. 2022. View Article : Google Scholar : PubMed/NCBI

66 

Lou T, Zhang L, Jin Z, Miao C, Wang J and Ke K: miR-455-5p enhances 5-fluorouracil sensitivity in colorectal cancer cells by targeting PIK3R1 and DEPDC1. Open Med (Wars). 17:847–856. 2022. View Article : Google Scholar : PubMed/NCBI

67 

Wilkinson L and Gathani T: Understanding breast cancer as a global health concern. Br J Radiol. 95:202110332022. View Article : Google Scholar : PubMed/NCBI

68 

Zhao H, Yu M, Sui L, Gong B, Zhou B, Chen J, Gong Z and Hao C: High expression of DEPDC1 promotes malignant phenotypes of breast cancer cells and predicts poor prognosis in patients with breast cancer. Front Oncol. 9:2622019. View Article : Google Scholar : PubMed/NCBI

69 

Zhang L, Du Y, Xu S, Jiang Y, Yuan C, Zhou L, Ma X, Bai Y, Lu J and Ma J: DEPDC1, negatively regulated by miR-26b, facilitates cell proliferation via the up-regulation of FOXM1 expression in TNBC. Cancer Lett. 442:242–251. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Sekhoacha M, Riet K, Motloung P, Gumenku L, Adegoke A and Mashele S: Prostate cancer review: Genetics, diagnosis, treatment options, and alternative approaches. Molecules. 27:57302022. View Article : Google Scholar : PubMed/NCBI

71 

Ramalho-Carvalho J, Martins JB, Cekaite L, Sveen A, Torres-Ferreira J, Graça I, Costa-Pinheiro P, Eilertsen IA, Antunes L, Oliveira J, et al: Epigenetic disruption of miR-130a promotes prostate cancer by targeting SEC23B and DEPDC1. Cancer Lett. 385:150–159. 2017. View Article : Google Scholar : PubMed/NCBI

72 

Badwelan M, Muaddi H, Ahmed A, Lee KT and Tran SD: Oral squamous cell carcinoma and concomitant primary tumors, what do we know? A review of the literature. Curr Oncol. 30:3721–3734. 2023. View Article : Google Scholar : PubMed/NCBI

73 

Qiu J, Tang Y, Liu L, Yu J, Chen Z, Chen H and Yuan R: FOXM1 is regulated by DEPDC1 to facilitate development and metastasis of oral squamous cell carcinoma. Front Oncol. 12:8159982022. View Article : Google Scholar : PubMed/NCBI

74 

Huang G, Chen S, Washio J, Paka Lubamba G, Takahashi N and Li C: Glycolysis-related gene analyses indicate that DEPDC1 promotes the malignant progression of oral squamous cell carcinoma via the WNT/β-catenin signaling pathway. Int J Mol Sci. 24:19922023. View Article : Google Scholar : PubMed/NCBI

75 

Pietras W: Advances and changes in the treatment of children with nephroblastoma. Adv Clin Exp Med. 21:809–820. 2012.PubMed/NCBI

76 

Geng G, Li Q, Guo X, Ni Q, Xu Y, Ma Z, Wang Y and Ming M: FOXO3a-modulated DEPDC1 promotes malignant progression of nephroblastoma via the Wnt/β-catenin signaling pathway. Mol Med Rep. 26:7272022. View Article : Google Scholar : PubMed/NCBI

77 

Geng G, Xu Y, Li Q, Li Q, Yuan L, Dong M and Ming M: S100A16 cooperates with DEPDC1 to promote the progression and angiogenesis of nephroblastoma through PI3K/Akt/mTOR pathway. Pol J Pathol. 74:182–193. 2023. View Article : Google Scholar : PubMed/NCBI

78 

Feng X, Zhang C, Zhu L, Zhang L, Li H, He L, Mi Y, Wang Y, Zhu J and Bu Y: DEPDC1 is required for cell cycle progression and motility in nasopharyngeal carcinoma. Oncotarget. 8:63605–63619. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Kikuchi R, Sampetrean O, Saya H, Yoshida K and Toda M: Functional analysis of the DEPDC1 oncoantigen in malignant glioma and brain tumor initiating cells. J Neurooncol. 133:297–307. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Yang M, Zhang H, Gao S and Huang W: DEPDC1 and KIF4A synergistically inhibit the malignant biological behavior of osteosarcoma cells through Hippo signaling pathway. J Orthop Surg Res. 18:1452023. View Article : Google Scholar : PubMed/NCBI

81 

Peng WX, Koirala P and Mo YY: LncRNA-mediated regulation of cell signaling in cancer. Oncogene. 36:5661–5667. 2017. View Article : Google Scholar : PubMed/NCBI

82 

Cui XY, Zhan JK and Liu YS: Roles and functions of antisense lncRNA in vascular aging. Ageing Res Rev. 72:1014802021. View Article : Google Scholar : PubMed/NCBI

83 

Yang L, Wu Y, Xu H, Zhang J, Zheng X, Zhang L, Wang Y, Chen W and Wang K: Identification and validation of a novel six-lncRNA-based prognostic model for lung adenocarcinoma. Front Oncol. 11:7755832022. View Article : Google Scholar : PubMed/NCBI

84 

Li N, Yu K, Huang D, Li S, Zeng D, Li J and Fan L: Molecular characterization of cuproptosis-related lncRNAs: Defining molecular subtypes and a prognostic signature of ovarian cancer. Biol Trace Elem Res. 202:1428–1445. 2024. View Article : Google Scholar : PubMed/NCBI

85 

Lu L, Liu LP, Zhao QQ, Gui R and Zhao QY: Identification of a ferroptosis-related LncRNA signature as a novel prognosis model for lung adenocarcinoma. Front Oncol. 11:6755452021. View Article : Google Scholar : PubMed/NCBI

86 

Xu W, Wang J, Xu J, Li S, Zhang R, Shen C, Xie M, Zheng B and Gu M: Long non-coding RNA DEPDC1-AS1 promotes proliferation and migration of human gastric cancer cells HGC-27 via the human antigen R-F11R pathway. J Int Med Res. 50:30006052210931352022. View Article : Google Scholar : PubMed/NCBI

87 

Mattick JS, Amaral PP, Carninci P, Carpenter S, Chang HY, Chen LL, Chen R, Dean C, Dinger ME, Fitzgerald KA, et al: Long non-coding RNAs: Definitions, functions, challenges and recommendations. Nat Rev Mol Cell Biol. 24:430–447. 2023. View Article : Google Scholar : PubMed/NCBI

88 

Arumugam P, Ramesh V, Sampathkumar B, Perumalsamy H, Balusamy SR, Suganya K, Balraj S, Nachimuthu SK and Sundaravadivelu S: Integrative transcriptome analysis of triple negative breast cancer profiles for identification of druggable targets. J Biomol Struct Dyn. 41:12106–12119. 2023. View Article : Google Scholar : PubMed/NCBI

89 

Wertz IE, Kusam S, Lam C, Okamoto T, Sandoval W, Anderson DJ, Helgason E, Ernst JA, Eby M, Liu J, et al: Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7. Nature. 471:110–114. 2011. View Article : Google Scholar : PubMed/NCBI

90 

Xiang Y, Zhang Q, Wei S, Huang C, Li Z and Gao Y: Paeoniflorin: A monoterpene glycoside from plants of Paeoniaceae family with diverse anticancer activities. J Pharm Pharmacol. 72:483–495. 2020. View Article : Google Scholar : PubMed/NCBI

91 

Xie L, Zhao YX, Zheng Y and Li XF: The pharmacology and mechanisms of platycodin D, an active triterpenoid saponin from Platycodon grandiflorus. Front Pharmacol. 14:11488532023. View Article : Google Scholar : PubMed/NCBI

92 

Nelde A, Rammensee HG and Walz JS: The peptide vaccine of the future. Mol Cell Proteomics. 20:1000222021. View Article : Google Scholar : PubMed/NCBI

93 

Obara W, Ohsawa R, Kanehira M, Takata R, Tsunoda T, Yoshida K, Takeda K, Katagiri T, Nakamura Y and Fujioka T: Cancer peptide vaccine therapy developed from oncoantigens identified through genome-wide expression profile analysis for bladder cancer. Jpn J Clin Oncol. 42:591–600. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Obara W, Eto M, Mimata H, Kohri K, Mitsuhata N, Miura I, Shuin T, Miki T, Koie T, Fujimoto H, et al: A phase I/II study of cancer peptide vaccine S-288310 in patients with advanced urothelial carcinoma of the bladder. Ann Oncol. 28:798–803. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Obara W, Hara I, Kato Y, Kato R, Inoue K, Sato F, Mimata H, Nakamura Y and Fujioka T: Immunotherapy with cancer peptides in combination with intravesical bacillus Calmette-Guerin for patients with non-muscle invasive bladder cancer. Cancer Immunol Immunother. 67:1371–1380. 2018. View Article : Google Scholar : PubMed/NCBI

96 

Fujiwara Y, Okada K, Omori T, Sugimura K, Miyata H, Ohue M, Kobayashi S, Takahashi H, Nakano H, Mochizuki C, et al: Multiple therapeutic peptide vaccines for patients with advanced gastric cancer. Int J Oncol. 50:1655–1662. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Fujiwara Y, Sugimura K, Miyata H, Omori T, Nakano H, Mochizuki C, Shimizu K, Saito H, Ashida K, Honjyo S, et al: A pilot study of post-operative adjuvant vaccine for advanced gastric cancer. Yonago Acta Med. 60:101–105. 2017. View Article : Google Scholar : PubMed/NCBI

98 

Daiko H, Marafioti T, Fujiwara T, Shirakawa Y, Nakatsura T, Kato K, Puccio I, Hikichi T, Yoshimura S, Nakagawa T, et al: Exploratory open-label clinical study to determine the S-588410 cancer peptide vaccine-induced tumor-infiltrating lymphocytes and changes in the tumor microenvironment in esophageal cancer patients. Cancer Immunol Immunother. 69:2247–2257. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Murahashi M, Hijikata Y, Yamada K, Tanaka Y, Kishimoto J, Inoue H, Marumoto T, Takahashi A, Okazaki T, Takeda K, et al: Phase I clinical trial of a five-peptide cancer vaccine combined with cyclophosphamide in advanced solid tumors. Clin Immunol. 166–167. 48–58. 2016.

100 

Jonker DJ, O'Callaghan CJ, Karapetis CS, Zalcberg JR, Tu D, Au HJ, Berry SR, Krahn M, Price T, Simes RJ, et al: Cetuximab for the treatment of colorectal cancer. N Engl J Med. 357:2040–2048. 2007. View Article : Google Scholar : PubMed/NCBI

101 

Kikuchi R, Ueda R, Saito K, Shibao S, Nagashima H, Tamura R, Morimoto Y, Sasaki H, Noji S, Kawakami Y, et al: A pilot study of vaccine therapy with multiple glioma oncoantigen/glioma angiogenesis-associated antigen peptides for patients with recurrent/progressive high-grade glioma. J Clin Med. 8:2632019. View Article : Google Scholar : PubMed/NCBI

102 

Tsuruta M, Ueda S, Yew PY, Fukuda I, Yoshimura S, Kishi H, Hamana H, Hirayama M, Yatsuda J, Irie A, et al: Bladder cancer-associated cancer-testis antigen-derived long peptides encompassing both CTL and promiscuous HLA class II-restricted Th cell epitopes induced CD4+ T cells expressing converged T-cell receptor genes in vitro. Oncoimmunology. 7:e14156872018. View Article : Google Scholar : PubMed/NCBI

103 

Tosi A, Dalla Santa S, Cappuzzello E, Marotta C, Walerych D, Del Sal G, Zanovello P, Sommaggio R and Rosato A: Identification of a HLA-A*0201-restricted immunogenic epitope from the universal tumor antigen DEPDC1. Oncoimmunology. 6:e13133712017. View Article : Google Scholar : PubMed/NCBI

104 

Yatsuda J, Irie A, Harada K, Michibata Y, Tsukamoto H, Senju S, Tomita Y, Yuno A, Hirayama M, Abu Sayem M, et al: Establishment of HLA-DR4 transgenic mice for the identification of CD4+ T cell epitopes of tumor-associated antigens. PLoS One. 8:e849082013. View Article : Google Scholar : PubMed/NCBI

105 

Slingluff CL Jr, Lee S, Zhao F, Chianese-Bullock KA, Olson WC, Butterfield LH, Whiteside TL, Leming PD and Kirkwood JM: A randomized phase II trial of multiepitope vaccination with melanoma peptides for cytotoxic T cells and helper T cells for patients with metastatic melanoma (E1602). Clin Cancer Res. 19:4228–4238. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2024
Volume 28 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu D, Li H and Ouyang J: Roles of DEPDC1 in various types of cancer (Review). Oncol Lett 28: 518, 2024.
APA
Liu, D., Li, H., & Ouyang, J. (2024). Roles of DEPDC1 in various types of cancer (Review). Oncology Letters, 28, 518. https://doi.org/10.3892/ol.2024.14651
MLA
Liu, D., Li, H., Ouyang, J."Roles of DEPDC1 in various types of cancer (Review)". Oncology Letters 28.5 (2024): 518.
Chicago
Liu, D., Li, H., Ouyang, J."Roles of DEPDC1 in various types of cancer (Review)". Oncology Letters 28, no. 5 (2024): 518. https://doi.org/10.3892/ol.2024.14651