Open Access

Targeting TRIP13 for overcoming anticancer drug resistance (Review)

  • Authors:
    • Liwen Zhao
    • Siyu Ye
    • Shengnan Jing
    • Yong-Jing Gao
    • Tianzhen He
  • View Affiliations

  • Published online on: September 29, 2023     https://doi.org/10.3892/or.2023.8639
  • Article Number: 202
  • Copyright: © Zhao 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

Cancer is one of the greatest dangers to human wellbeing and survival. A key barrier to effective cancer therapy is development of resistance to anti‑cancer medications. In cancer cells, the AAA+ ATPase family member thyroid hormone receptor interactor 13 (TRIP13) is key in promoting treatment resistance. Nonetheless, knowledge of the molecular processes underlying TRIP13‑based resistance to anticancer therapies is lacking. The present study evaluated the function of TRIP13 expression in anticancer drug resistance and potential methods to overcome this resistance. Additionally, the underlying mechanisms by which TRIP13 promotes resistance to anticancer drugs were explored, including induction of mitotic checkpoint complex surveillance system malfunction, promotion of DNA repair, the enhancement of autophagy and the prevention of immunological clearance. The effects of combination treatment, which include a TRIP13 inhibitor in addition to other inhibitors, were discussed. The present study evaluated the literature on TRIP13 as a possible target and its association with anticancer drug resistance, which may facilitate improvements in current anticancer therapeutic options.

Introduction

Thyroid hormone receptor interactor 13 (TRIP13) is a member of the AAA+ ATPase family able to generate mechanical stresses through ATP hydrolase activities (1). TRIP13 was initially identified as a protein that interacts with the E1 protein of the human papillomavirus (2). In 1999, TRIP13 was found to be required for the meiotic checkpoint in yeast (3). Studies have showed involvement of TRIP13 in meiotic recombination and DNA repair in several organisms, including rice (4), yeast (5), Drosophila melanogaster (6), Caenorhabditis elegans (7) and mice (810). Additionally, it has been identified as a constituent of the spindle assembly checkpoint (SAC) pathway (1113), which is involved in the accurate segregation of chromosomes (14). TRIP13 [and its homolog pachytene checkpoint 2 (PCH2) is an AAA+ ATPase that produces homohexamers and uses ATP as a substrate (3,8,9, 1519). PCH2 binds to Hop1 and alters its structure, which displaces Hop1 from DNA (20). Hydrolysis of ATP by TRIP13/PCH2 provides energy necessary to undergo conformational changes that exert mechanical force on Hop1 (21). In addition to its role as a kinetochore protein, TRIP13/PCH2 interacts with the p31comet protein, which is responsible for gene silencing (11). TRIP13/PCH2 is linked to various malignancies due to its involvement in ensuring proper biorientation of chromosomes during mitosis; TRIP13 has been shown to be overexpressed in a number of malignancies, including colorectal (2224), head and neck (25), breast (26,27), lung (2830), liver (31,32) and prostate cancer (33,34), multiple myeloma (35), bladder cancer (36,37) and human chronic lymphoblastic leukemia (38). Importantly, overexpression of TRIP13 promotes advancement of head and neck squamous cell carcinoma (HNSCC) (25) and lung adenocarcinoma (39). Depletion of TRIP13 or suppression of its activity has been demonstrated to diminish tumor development in head and neck and colon cancer and hepatocellular carcinoma (HCC) (32,40). TRIP13 also serves an essential role in the survival and spread of tumor stem cells in cutaneous melanoma (41), prostate cancer (33) and lung adenocarcinoma (39). Based on these findings, TRIP13 serves a key role in tumor development.

Effective cancer therapy is hampered significantly by the development of resistance to anticancer medications. Overexpression of TRIP13 has been linked to decreased sensitivity to anticancer medicines (such as bortezomib and cisplatin) (25,42). TRIP13 facilitates development of nedaplatin resistance in esophageal squamous cell carcinoma (43). In addition, synergistic anti-HCC efficacy is achieved by mixing TRIP13 inhibitor DCZ0415 with the PARP1 inhibitor olaparib (44). Therefore, TRIP13 is involved in drug resistance in cancer cells. The present study evaluated the function of TRIP13 expression in anticancer drug resistance and potential methods to overcome this resistance. Furthermore, the present study explored the underlying mechanism of targeting TRIP13 to overcome anticancer drug resistance and summarized the roles TRIP13 plays in cancer treatment. The present study also reviewed the effects of combination treatment, which include a TRIP13 inhibitor in addition to other inhibitors.

Structure and biological functions of TRIP13

The structure of human TRIP13, ATP-bound form (45) is shown in Fig. 1. Additionally, TRIP13/PCH2 exhibits kinetochore protein activity by interacting with the silencing protein p31comet (11).

TRIP13/PCH2 has been shown to have a role in mitosis, namely in the transition from metaphase to anaphase, as well as the SAC (19). It also releases anaphase-promoting complex (APC) from checkpoint inhibition (19). Before anaphase, the cell must make sure its chromosomes are appropriately organized and bioriented for separation of sister chromatids. Numerous proteins such as spindle checkpoint, securin and cyclin B, are needed for this process to maintain accurate timing and reliable separation. The activation of the APC is required for mitosis. Protein CDC20, which is typically suppressed by the mitotic checkpoint complex (MCC), activates the APC. The TRIP13-related gene Mad2 exists in two isoforms, the open (O-) form and the closed (C-) form (19). O-Mad2 changes into C-Mad2 when kinetochores detach and C-Mad2 may then hook onto CDC20 and sequester it, blocking mitotic progression (46). MCC must be disassembled, and this process is mediated by p31comet (47). This is hypothesized to take place in part via structural mimicry as p31comet has structural similarities to C-Mad2 (48). Nevertheless, ATP is needed for this step. TRIP13 uses p31comet as an adaptor protein to convert C-Mad2 into O-Mad2, then induces activation of the SAC and the formation of the MCC. To conclude, TRIP13/PCH2 is key for SAC activation and MCC formation.

Similarly, TRIP13/PCH2 is also involved in the G2/prophase stage of meiosis (3). TRIP13 affects the frequency of double-strand breaks (DSBs) promoting homology-directed repair, while suppressing non-homologous end joining and translesion synthesis. Homologous recombination following these breaks needs a protein complex to shape and direct the correct pairing of chromosomes. PCH2 gene in budding yeast is necessary for the meiotic checkpoint that inhibits chromosomal segregation when recombination and chromosome synapsis are impaired (3). TRIP13 is necessary for the creation of the synaptonemal complex (SC), which that builds chromosome pairs. In meiocytes lacking TRIP13, pericentric synaptic forks are more common, crossovers are reduced and chiasma (the site of contact between homologous chromosomes) distribution is disrupted (9). SC synthesis necessitates the elimination of HORMA domain family proteins (HORMADs) during meiosis. PCH2 is required for SC formation to remove Hop1 from chromosomes (49). In mouse spermatocytes, TRIP13 facilitates depletion of other HORMADs, including HORMAD1 and HORMAD2 (10). These findings demonstrate a key dynamic function for TRIP13/PCH2 in removal of several proteins during SC formation and subsequent meiosis.

Implications of TRIP13 in cancer

HNSCC exhibits TRIP13 overexpression, which promotes proliferation and invasion (25). Overexpression of both TRIP13 and Mad2 are associated with various types of cancer, including multiple myeloma (42), head and neck cancer (25), colorectal cancer (2224), chronic lymphocytic leukemia (50), lung adenocarcinoma (39) and prostate cancer (33). Overexpression of TRIP13 attenuates the mitotic delay caused by Mad2 overexpression, but downregulation of TRIP13 compounds the effects of Mad2 overexpression. In addition, downregulating TRIP13 and overexpressing Mad2 suppresses proliferation in cells and tumor xenografts, suggesting therapeutic potential for inhibition of TRIP13 (47). Mad2 promotes drug resistance in ovarian cancer (51). The expression of TRIP13 is markedly increased in epithelial ovarian cancer (EOC) cell lines (SKOV-3, HEY and OVCAR-3) compared with normal ovarian cell lines (52). Additionally, knockdown of TRIP13 in EOC cells inhibits cell proliferation, decreases cell invasion and migration and stimulates apoptosis (52). This demonstrates that TRIP13 promotes the development of tumor cells and may be a potential target for tumor therapy.

TRIP13 and anticancer drug resistance

Anticancer regimens have the potential to kill most cancer cells at first, but some malignant cells survive because they have either already developed mechanisms of drug resistance (intrinsic resistance) or have acquired them through random mutation and genetic alteration (acquired resistance). Resistance mechanisms include: Decreased expression or dysfunction of influx drug transporters, increased activity of multidrug-resistant (MDR) efflux pumps of the ATP-binding cassette (ABC) superfamily, such as P-glycoprotein, multidrug resistance associated protein 1 (MRP1) and breast cancer resistance protein (ABCG2) (53), qualitative and quantitative changes in the drug target and drug sequestration within intracellular compartments. MDR efflux pumps remove numerous anticancer medications from cancer cells or store them in organelles such as lysosomes, where they are inaccessible to the cell target sites (54). The emergence of resistance to anticancer drugs is associated with genomic instability, which may include mutations, amplifications, deletion and/or translocations (55). MDR is caused by a combination of variables, including genetics (gene mutations, amplification and epigenetic modifications), growth hormones and higher DNA repair ability (56). According to Vasan et al (57), the primary factors that determine treatment resistance include: Tumor burden and growth dynamics; tumor heterogeneity; physical barriers; the immune system and the tumor microenvironment. All of these factors contribute to the diminished effectiveness of the medications used to treat the tumor.

TRIP13 expression is shown to be higher in samples from patients with multiple myeloma compared with control samples (42,58). Overexpression of TRIP13 is linked to decreased sensitivity to bortezomib and cisplatin (25,42,59). TRIP13 is shown to be responsible for the development of nedaplatin resistance in esophageal squamous cell carcinoma (43). Furthermore, considering the role of cisplatin (6062) and PARP (6365) inhibitors in the treatment of ovarian cancer, further study is needed to elucidate the role of TRIP13 in treatment-induced drug resistance caused by cisplatin and PARP therapy. These aforementioned results suggest a connection between elevated TRIP13 levels and drug resistance.

Targeting TRIP13 for overcoming anticancer drug resistance

Targeting TRIP13 is a potential strategy for overcoming drug resistance. Inhibiting TRIP13 makes HNSCC cells more sensitive to effects of radiation and chemotherapy (25). Recent research found that TRIP13−/− HCC cells are more vulnerable to the effects of chemotherapy than normal HCC cells (44). DCZ0415 may stimulate antimyeloma activity in primary cells generated from individuals with myeloma who are resistant to several drugs (35). Inhibition or depletion of TRIP13 may thus constitute a viable approach to circumventing resistance that anticancer drugs cause.

Underlying mechanism of TRIP13-mediated anticancer drug resistance

There are primarily three molecular mechanisms involved in TRIP13-mediated drug resistance. These mechanisms include promoting chromosomal instability, enhancing autophagy and modulating the tumor microenvironment (Fig. 2).

TRIP13 promotes drug resistance by causing chromosome instability

Previous studies have suggested that SAC is a universal safeguard that guarantees the integrity of chromosomal separation in cell division (6668). Multiple malignancies exhibit overexpression of SAC proteins, which is linked to chromosome instability (CIN) in tumors (6971). Additionally, telomere dysfunction (7274) and faulty DNA repair mechanism response (75) contribute significantly to CIN in cancer. Not only does CIN serve a role in the origin, maintenance and growth of tumors, but it also stimulates the development of treatment resistance in cancer cells. TRIP13 is associated with CIN in human malignancies such as multiple myeloma (12,76,77) and treatment resistance. Previous studies have documented that TRIP13 plays a role in the induction of CIN and drug resistance via SAC signaling and modulating DNA damage repair (25,42,43,58,78).

TRIP13 induces drug resistance via inducing the dysfunction of MCC surveillance system

MCC, also known as SAC effector (79), is made of the proteins Mad2, BubR1/Mad3 and Bub3, in addition to CDC20 (68,80,81). Mis-segregation of chromosomes and failure to stop in mitosis may result from malfunctions in the MCC monitoring system. Both of these contribute to the formation of human tumors as well as medication resistance in cancer (82). Subcellular localization research demonstrates that TRIP13 co-localizes with Mad2 at kinetochores and interacts with p31comet, a Mad2-binding HORMA-domain protein that negatively controls SAC localization to kinetochores in prometaphase. When TRIP13 is overexpressed, Mad2 protein levels are reduced (42). Mad2 forms a compound with APC/cyclosome (APC/C) when the MCC monitoring system is activated. This prevents degradation of securin and cyclin B1, which ultimately leads to arrest of the cells in the prometaphase phase of the cell cycle (83). Resistance to paclitaxel is associated with a weaker spindle checkpoint in conjunction with lower expression of Mad2 (84). Through activation of the Akt signaling cascade, TRIP13 ubiquitinates and degrades the checkpoint surveillance protein Mad2, which further results in damaged checkpoint surveillance and subsequent drug resistance (42). Inhibition of PI3k/Akt may partially rescue TRIP13-induced drug resistance to bortezomib (42). The aforementioned studies indicate that TRIP13/Mad2 axis, via activating PI3K-Akt signaling pathway, leads to damage to the checkpoint surveillance system and subsequent anticancer drug resistance. Hence, TRIP13-induced anti-apoptosis action and dysfunction in MCC surveillance system contribute to chromosome instability, leading to drug resistance in cancer cells.

TRIP13 induces drug resistance by promoting DNA repair

To prevent genomic instability in the host, DNA damage response and repair mechanisms have been conserved throughout evolution (85) of both prokaryotes and eukaryotes. In mammalian cells, dysregulation of proteins involved in these processes may increase genomic changes, which lead to genomic instability, a well-established hallmark of cancer (86,87). There has been emergence of new and promising techniques for targeting the DNA damage response and repair pathways to increase cancer cell sensitivity to existing therapeutic drugs (88). Targeting DNA damage is signaling and repair may interrupt the compensatory activation of DNA repair pathways that may function as a drug resistance mechanism. For example, targeting DNA repair has become a legitimate therapeutic approach. This method uses PARP inhibitors to treat breast, ovarian, pancreatic and prostate cancers that have DNA repair deficiencies (8995). Clinical research and development of small compounds that target key components of the DNA damage response and repair pathways, including DNA-dependent protein kinase (DNA-PK), ataxia telangiectasia mutated (ATM) and Rad3-related kinase (ATR), ATM and checkpoint kinase 1 (CHK1), have been accelerated (96). In addition, accurate targeting of these key molecules offers the possibility of using biomarkers of DNA repair deficiency to choose the most effective therapy for each individual patient to achieve the highest possible therapeutic index (96).

Recent research has provided support for the hypothesis that TRIP13-induced anti-apoptosis may contribute to drug resistance in cancer cells (97). This is because one of the primary strategies anticancer medications utilize to trigger cell death is induction of apoptosis. When TRIP13 is overexpressed, cancer cells are less sensitive to bortezomib and cisplatin (25,42). When multiple myeloma cells transfected with TRIP13 are treated with bortezomib and etoposide, cell viability experiment indicates that the number of viable tumor cells is greater in the TRIP13-transfected cells compared with control cells (42). Comparatively, HNSCC cells that overexpress TRIP13 display a decreased sensitivity to cisplatin when compared with control cells (25). Therefore, TRIP13 plays a role in the development of drug resistance in cancer cells. When treated with increasing doses of bortezomib, TRIP13-overexpressing ARP1 and OCIMy5 multiple myeloma cell lines are less likely to undergo apoptosis and better able to withstand the cytotoxic effects compared with cells transfected with empty vectors. In TRIP13-overexpressing ARP1 multiple myeloma cell line, the G2/M cell cycle arrest induced by bortezomib is consistently suppressed compared with control cells (42). In addition, TRIP13 small interfering (si)RNA knockdown in multiple myeloma cells eliminates doxycycline treatment resistance and triggers apoptosis both in vitro and in a mouse model of xenograft myeloma (35,42). In ARP1 and OCIMy5 multiple myeloma cells, downregulation of TRIP13 leads to an increase in levels of cleaved PARP and activation of caspase 3, both of which indicate a potential role for knockdown of TRIP13 in inhibiting the apoptotic pathway (42). Similarly, in human chronic lymphocytic leukemia, microarray data evaluated using the ‘canonical pathway’ module of Ingenuity route analysis reveal that TRIP13 participates in numerous apoptosis-associated pathways, including ‘induction of apoptosis by HIV1’, ‘p53 signaling’ and ‘PPAR signaling’ (50). In addition, knocking down TRIP13 causes a notable increase in the activity of caspase 3/7 in Granta-519 and JVM-2 B cell lymphocytic leukemia cell lines (50). The modulation of the c-Myc/TRIP13/p53 upregulated modulator of apoptosis (PUMA) axis is the mechanism by which TRIP13 serves a role in the development of chronic lymphocytic leukemia (50). In HNSCC, suppression of TRIP13 also induces cell cycle arrest (25). In cells transfected with TRIP13 siRNA, there is a greater accumulation of phosphorylated histone H2A histone family member X (the marker of DSBs) (25). According to western blot analysis, DSBs caused by TRIP13 siRNA occur prior to apoptosis (25). These results provided compelling evidence that TRIP13 increase DNA repair, which in turn leads to treatment resistance.

TRIP13 induces drug resistance by enhancing autophagy

Autophagy is a natural method of cell survival that is effectively employed by tumor cells to prevent cell death and generate drug resistance (98100). Both of these may be accomplished by tumor cells via autophagy, a macromolecular process in which cells break down and recycle intracellular substrates and damaged organelles to reduce cell stress caused by factors such as nutritional deficiency, hypoxia, irradiation and cytotoxic chemicals. When cancer is in the early stages, autophagy has been shown to protect against malignant disease; nevertheless, transformed cells exhibit increased autophagy to promote survival, proliferation and metastasis (101103). Although the precise function of autophagy in cell death and survival remains unclear, autophagy is enhanced in cancer cells exposed to stressful situations, such as anticancer treatment, which may result in anticancer drug resistance (104106). Previous research examined TRIP13 role in autophagy by treating cells with DMSO (control), gefitinib (an autophagy agonist) and 3-methyladenine (3-MA; an autophagy inhibitor) (107); TRIP13 induces autophagy in non-small cell lung cancer (NSCLC) cells, shown by an increase in the number of LC3B (autophagy marker)-positive puncta seen by immunofluorescence examination (107). Overexpression of TRIP13 in NSCLC cells leads to an increase in LC3B and decrease in autophagy marker P62 expression (107). The opposite effects are seen in tumor cells when TRIP13 expression is suppressed (107). All of the aforementioned effects are mitigated by the gefitinib therapy, but 3-MA has the opposite effect (107). Therefore, TRIP13 may be responsible for inducing gefitinib resistance in NSCLC cells via increasing autophagy (107). Therefore, TRIP13 can be used as a biomarker and therapeutic target autophagy for overcoming drug resistance.

TRIP13 induces drug resistance by preventing immune clearance

The tumor microenvironment, which comprises immune cells, stroma and vasculature, may promote drug resistance via numerous methods, including inhibiting immune clearance of tumor cells, preventing drug absorption and increasing paracrine growth factors, to promote cancer cell development (108). DCZ0415 prevents proliferation inhibition of multiple myeloma cells even in the presence of bone marrow stromal cells and the cytokines IL6 and IGF1 in a cellular experiment that mimics multiple myeloma in its microenvironment (35). In addition to cytotoxicity against multiple myeloma cells, DCZ0415 also targets the bone marrow microenvironment and overcomes the proliferative effects on bone marrow stromal cells (35).

Additionally, suppression of TRIP13 stimulates an anticancer immune response by increasing production of cytotoxic mediators. Recent studies show that TRIP13 inhibition promotes the invasion of CD3+, CD4+ and CD8+ T cells (35). DCZ0415 considerably increases the production of cytotoxic mediators such as granzyme B, perforin and IFN-γ, which may contribute to the cytotoxic effect against murine MC38 cells. In addition, blocking immunological checkpoints, such as PD-1 and cytotoxic T-lymphocyte-associated protein 4 (CTLA4), in tumors treated with DCZ0415 has the potential to increase cytotoxicity and regression of the tumor (109). Hence, decreased expression of TRIP13 stimulates immune responses in the microenvironment of the tumor, which may help overcome drug resistance to anticancer medications.

Combination therapy for overcoming anticancer drug resistance

DNA DSBs and DNA damage responses induced by certain anticancer drugs, such as example, β-emitter iodine-131, O-6-methylguanine-DNA methyltransferases and cisplatin, make cancer cells more vulnerable to further treatment (110). DNA damage response followed by effective repair of DSBs is key to maintain genomic integrity. On the other hand, in cancer, the repair of anticancer agent-induced DSBs via the non-homologous end joining (NHEJ) or homologous recombination (HR) repair pathways enhances treatment resistance and recurrence in patients (111). According to recent research, TRIP13 improves NHEJ repair and generates treatment resistance in head and neck cancer via binding to NHEJ proteins Ku protein with molecular weight of 70 KDa (KU70, encoded by the X-ray repair cross-complementing protein 6 gene located on chromosome 22), Ku protein with molecular weight of 80 KDa, (KU80,encoded by the X-ray repair cross-complementing protein 5 gene on chromosome 2) and DNA-dependent protein kinase catalytic subunit (DNA-PKcs) (25). Additionally, GFP-based reporter experiment demonstrates that DCZ0415 inhibits DNA repair by the NHEJ repair pathway (35). Hence, combining TRIP13 inhibitor with other inhibitors may be an option to overcome anticancer drug resistance (Table I).

Table I.

Combination of TRIP13 and TRIP13 depletion inhibitors and other drugs.

Table I.

Combination of TRIP13 and TRIP13 depletion inhibitors and other drugs.

TreatmentTRIP13 effectPharmacological interactionTumorFirst author (year)(Refs.)
DCZ0415 with PARP1 inhibitor OlaparibImpaired DNA repair gene regulation and expressionSynergisticHepatocellular CarcinomaXu et al (2022)[44] J Cancer (2022) 13 (7): 2226–2237.
Combining Aurora kinase inhibition with TRIP13 depletionLed to extensive apoptosisSynergisticHPV-negative cancer cellsSoma Ghosh et al (2022)[97] Clin Cancer Res (2022) 28 (20): 4479–4493.
DCZ0415 with the multiple myeloma chemotherapeutic melphalanImpaired nonhomologous end joining repair and inhibited NF-κB activitySynergisticMultiple MyelomaYingcong Wang et al (2020)[35] Cancer Res (2020) 80 (3): 536–548.
DCZ0415 with the HDAC inhibitor PanobinostatImpaired nonhomologous end joining repair and inhibited NF-κB activitySynergisticMultiple MyelomaYingcong Wang et al (2020)[35] Cancer Res (2020) 80 (3): 536–548.
Combining the DNA-PKcs inhibitor Nu7026 with TRIP13 depletionTRIP13 provides the energy needed for assembly of the DNA-PKcs complexSynergisticHead and neck cancerRajat Banerjee et al (2014)[25] Nat Commun. 2014; 5: 4527.

[i] TRIP13, thyroid hormone receptor interactor 13; DCZ0415, a small-molecule inhibitor targeting TRIP13; PARP1, poly(ADP-ribose) polymerase 1; Olaparib, an inhibitor of the enzyme poly ADP ribose polymerase (PARP); DNA, deoxyribonucleic acid; HPV, Human papillomavirus; NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells; HDAC, Histone deacetylases; DNA-PKcs, DNA-dependent protein kinase, catalytic subunit; Nu7026, a potent DNA-PK inhibitor

In addition, the upregulation of the TRIP13-mediated NHEJ repair pathway is the cause of the resistance to anticancer drugs (such as bortezomib and cisplatin). Therefore, cancer cells that have high levels of TRIP13 expression will be more susceptible to DNA-PKcs inhibitor than cancer cells with low levels of TRIP13 expression (25). Banerjee et al (25) showed that tumor cells that overexpress TRIP13 are more sensitive to the DNA-PKcs inhibitor Nu7026 (25). Hence, combining the TRIP13 inhibitor with DNA-PKcs inhibitor may be an effective treatment approach for TRIP13-mediated drug resistance.

Synergistic effects of DCZ0415 and PARP1 inhibitors

Anticancer drug resistance is partly attributed to TRIP13-induced DNA damage (25). Therefore, combining TRIP13 inhibitor with inhibitors against DNA repair proteins may overcome drug resistance (25). Synergistic effects of DCZ0415 and olaparib on increasing anticancer activity have been demonstrated in HCC cells (44). Specifically, in HCC cell lines, combination groups treated with DCZ0415 and olaparib show greater growth suppression compared with single-treatment groups, suggesting that this strategy may represent a viable therapeutic option for HCC (44). Furthermore, Clairmont et al (78) and Sarangi et al (112) demostrate that BRCA1−/− cells overexpressing TRIP13 or p31comet develop resistance to the PARP inhibitor Olaparib, and this resistance results from the reactivation of homologous recombination (HR)-mediated DNA repair owing to decreased Shieldin complex levels (113). Therefore, for individuals with BRCA1-deficient malignancies who acquire resistance to PARP medicines, blocking TRIP13/p31comet-mediated Shieldin disassembly may be a viable therapy option.

Synergistic effects of TRIP13 depletion and aurora kinase inhibitors

Ghosh et al (97) show low Rb levels drive sensitivity to aurora kinase inhibitors in human papillomavirus (HPV)-positive cancers (97). Mitotic kinases, in particular, aurora B (114) and A (114), are essential for maintenance of tumors that have loss-of-function RB1 mutations. Inhibition of Rb pathway activity upregulates MCC gene expression, which may lead to chromosomal instability and prolonged mitotic phase of the cell cycle (77,116). In patients with HNSCC, the expression of Bub1B and mitotic arrest deficient 2-like 1 (MAD2L1) is greater in HPV-positive tumors than in HPV-negative tumors. Depletion of Bub1B, as revealed by Gong et al (113), partly protects Rb-deficient SCLC cells from apoptosis produced by aurora kinase A inhibition. In a similar manner, knockdown of MAD2L1 and Bub1B significantly decreases the sensitivity to aurora kinase inhibitors in treatment of HPV-positive squamous carcinoma (97), while MAD2L1 regulator TRIP13 is depleted, which leads to an increase in the sensitivity to aurora kinase inhibitors for the treatment of HPV-positive cancers. If mitotic checkpoint gene MAD2L1 is overexpressed in HPV-positive cells and these cells need TRIP13 and aurora kinase activity to maintain mitotic fidelity, combined suppression of TRIP13 and aurora kinase activity may cause cancer cells to undergo irreversible mitotic arrest, DNA damage and death. According to previous research, the combination of TRIP13 depletion and aurora kinase inhibition leads to increased apoptosis compared with specific inhibition of a single route in HPV-positive tumors (97). Therefore, overexpression of TRIP13 makes cancer cells resistant to aurora kinase inhibitors, which are used in the treatment of HPV-positive squamous carcinoma. This resistance is induced by cancer cell ability to retain their mitotic fidelity. The combined therapeutic benefits of inhibiting TRIP13 and aurora kinase may be a potential technique for treating individuals with malignancies caused by Rb pathway abnormalities.

Discussion

TRIP13/PCH2 is associated with a number of malignancies as it ensures proper alignment of chromosomes during mitosis. As a result, TRIP13 may be a target for the development of an anticancer medication. In addition, overexpression of TRIP13 is associated with decreased susceptibility to bortezomib and cisplatin (25,42). In esophageal squamous cell carcinoma, TRIP13 causes nedaplatin resistance (43). In addition, combination of DCZ0415 with olaparib results in a synergistic effect on the activity of HCC (44). Thus, TRIP13 serves a role in cancer cell drug resistance.

To determine how TRIP13 contributes to the development of anticancer drug resistance, it is vital to research the mechanism that controls the regulation of TRIP13 expression. In perihilar cholangiocarcinoma cells, c-Myc stimulates the transcription of TRIP13 (117). In addition, transcription factor specificity protein 1 (SP1) serves a role in controlling the amount of TRIP13 that is expressed (44). TRIP13 expression is drastically reduced by SP1 inhibition or knockdown, whereas overexpression of SP1 notably increases TRIP13 expression (44). However, it is unclear whether c-Myc or SP1 are involved in development of TRIP13-mediated drug resistance. Because of this, identifying the transcription factors that govern expression of TRIP13 is required, as well as genes controlled by TRIP13, which may to act as targets for the development of cancer treatment.

Numerous studies (34,118120) have shown that microRNAs (miRNAs) directed against TRIP13 suppress or increase proliferation of cancer cells. Cancer may be treated using miRNA mimics or inhibitors, which can also circumvent resistance to anticancer medications (121). To facilitate development of new treatment regimens, for example, the combination of TRIP13 and immune check point inhibitors, it is necessary to have a better understanding of the effects of miRNAs and TRIP13-mediated drug resistance on immune responses in the tumor microenvironment.

To create anticancer treatments that are effective for people with cancer with high levels of TRIP13, it is necessary to identify new TRIP13-binding proteins and the TRIP13 binding domain. There is a possibility that peptides with a structure identical to the binding domain of TRIP13 may facilitate overcoming resistance to anti-cancer medications. For example, TRIP13 binds to ubiquitin-specific protase-7 (USP7), then TRIP13-induced resistance to proteasome inhibition can be overcome by a USP7 inhibitor (25,42,59).

Additionally, the microenvironment of a tumor comprises innate and adaptive immune cells (B and T, dendritic and myeloid-derived suppressor cells and M1/2 macrophages), as well as cancer cells and endothelial cells and cancer-associated fibroblasts. The tumor microenvironment is essential for the development and progression of cancer. There is evidence that interactions between cancer cells and immune cells may enhance the growth of cancer cells (108). Depletion of TRIP13 facilitates activation of immune responses in the tumor microenvironment (35), which improves anticancer treatment resistance. In immune cells, it is important to discover the genes controlled by the TRIP13 protein to develop potential immunotherapeutic targets.

TRIP13 inhibitor resistance may develop in future. Because of this, it will be required to identify genes that confer resistance to TRIP13 inhibitors as well as genes controlled by TRIP13 inhibitors. It is possible that immune evasion, drug efflux, the overexpression of oncogenes and the downregulation of tumor suppressor genes mediate resistance to TRIP13 inhibitors.

Acknowledgements

The authors would like to thank Dr Peng Zhao (Universiti Pendidikan Sultan Idris, Tanjong Malim, Malaysia) for critical review of manuscript.

Funding

The present study was supported by National Natural Science Foundation of China (grant no. 82100557), Science Foundation of Nantong (grant no. JC2021018), and Large Instruments Open Foundation of Nantong University (grant no. KFJN2375).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LZ, NJ and SY wrote the manuscript. TH and YJG performed the literature review and edited the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

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 

Jeong H, Wie M, Baek IJ, Sohn G, Um SH, Lee SG, Seo Y, Ra J, Lee EA, Kim S, et al: TRIP13 participates in immediate-early sensing of DNA strand breaks and ATM signaling amplification through MRE11. Cells. 11:40952022. View Article : Google Scholar : PubMed/NCBI

2 

Lee JW, Choi HS, Gyuris J, Brent R and Moore DD: Two classes of proteins dependent on either the presence or absence of thyroid hormone for interaction with the thyroid hormone receptor. Mol Endocrinol. 9:243–254. 1995. View Article : Google Scholar : PubMed/NCBI

3 

San-Segundo PA and Roeder GS: Pch2 links chromatin silencing to meiotic checkpoint control. Cell. 97:313–324. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Miao C, Tang D, Zhang H, Wang M, Li Y, Tang S, Yu H, Gu M and Cheng Z: Central region component1, a novel synaptonemal complex component, is essential for meiotic recombination initiation in rice. Plant Cell. 25:2998–3009. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Farmer S, Hong EJ, Leung WK, Argunhan B, Terentyev Y, Humphryes N, Toyoizumi H and Tsubouchi H: Budding yeast Pch2, a widely conserved meiotic protein, is involved in the initiation of meiotic recombination. PLoS One. 7:e397242012. View Article : Google Scholar : PubMed/NCBI

6 

Joyce EF and McKim KS: Drosophila PCH2 is required for a pachytene checkpoint that monitors double-strand-break-independent events leading to meiotic crossover formation. Genetics. 181:39–51. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Ye Q, Rosenberg SC, Moeller A, Speir JA, Su TY and Corbett KD: TRIP13 is a protein-remodeling AAA+ ATPase that catalyzes MAD2 conformation switching. Elife. 4:e073672015. View Article : Google Scholar : PubMed/NCBI

8 

Li XC and Schimenti JC: Mouse pachytene checkpoint 2 (trip13) is required for completing meiotic recombination but not synapsis. PLoS Genet. 3:e1302007. View Article : Google Scholar : PubMed/NCBI

9 

Roig I, Dowdle JA, Toth A, de Rooij DG, Jasin M and Keeney S: Mouse TRIP13/PCH2 is required for recombination and normal higher-order chromosome structure during meiosis. PLoS Genet. 6:e10010622010. View Article : Google Scholar : PubMed/NCBI

10 

Wojtasz L, Daniel K, Roig I, Bolcun-Filas E, Xu H, Boonsanay V, Eckmann CR, Cooke HJ, Jasin M, Keeney S, et al: Mouse HORMAD1 and HORMAD2, two conserved meiotic chromosomal proteins, are depleted from synapsed chromosome axes with the help of TRIP13 AAA-ATPase. PLoS Genet. 5:e10007022009. View Article : Google Scholar : PubMed/NCBI

11 

Tipton AR, Wang K, Oladimeji P, Sufi S, Gu Z and Liu ST: Identification of novel mitosis regulators through data mining with human centromere/kinetochore proteins as group queries. BMC Cell Biol. 13:152012. View Article : Google Scholar : PubMed/NCBI

12 

Wang K, Sturt-Gillespie B, Hittle JC, Macdonald D, Chan GK, Yen TJ and Liu ST: Thyroid hormone receptor interacting protein 13 (TRIP13) AAA-ATPase is a novel mitotic checkpoint-silencing protein. J Biol Chem. 289:23928–23937. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Eytan E, Wang K, Miniowitz-Shemtov S, Sitry-Shevah D, Kaisari S, Yen TJ, Liu ST and Hershko A: Disassembly of mitotic checkpoint complexes by the joint action of the AAA-ATPase TRIP13 and p31(comet). Proc Natl Acad Sci USA. 111:12019–12024. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Silva RD, Mirkovic M, Guilgur LG, Rathore OS, Martinho RG and Oliveira RA: Absence of the spindle assembly checkpoint restores mitotic fidelity upon loss of sister chromatid cohesion. Curr Biol. 28:2837–2844.e2833. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Bhalla N and Dernburg AF: A conserved checkpoint monitors meiotic chromosome synapsis in Caenorhabditis elegans. Science. 310:1683–1686. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Börner GV, Barot A and Kleckner N: Yeast Pch2 promotes domainal axis organization, timely recombination progression, and arrest of defective recombinosomes during meiosis. Proc Natl Acad Sci USA. 105:3327–3332. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Joshi N, Barot A, Jamison C and Börner GV: Pch2 links chromosome axis remodeling at future crossover sites and crossover distribution during yeast meiosis. PLoS Genet. 5:e10005572009. View Article : Google Scholar : PubMed/NCBI

18 

Vader G, Blitzblau HG, Tame MA, Falk JE, Curtin L and Hochwagen A: Protection of repetitive DNA borders from self-induced meiotic instability. Nature. 477:115–119. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Vader G: Pch2(TRIP13): Controlling cell division through regulation of HORMA domains. Chromosoma. 124:333–339. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Chen C, Jomaa A, Ortega J and Alani EE: Pch2 is a hexameric ring ATPase that remodels the chromosome axis protein Hop1. Proc Natl Acad Sci USA. 111:E44–E53. 2014.PubMed/NCBI

21 

Yedidi RS, Wendler P and Enenkel C: AAA-ATPases in protein degradation. Front Mol Biosci. 4:422017. View Article : Google Scholar : PubMed/NCBI

22 

Sheng N, Yan L, Wu K, You W, Gong J, Hu L, Tan G, Chen H and Wang Z: TRIP13 promotes tumor growth and is associated with poor prognosis in colorectal cancer. Cell Death Dis. 9:4022018. View Article : Google Scholar : PubMed/NCBI

23 

Kurita K, Maeda M, Mansour MA, Kokuryo T, Uehara K, Yokoyama Y, Nagino M, Hamaguchi M and Senga T: TRIP13 is expressed in colorectal cancer and promotes cancer cell invasion. Oncol Lett. 12:5240–5246. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Agarwal S, Behring M, Kim HG, Chandrashekar DS, Chakravarthi BVSK, Gupta N, Bajpai P, Elkholy A, Al Diffalha S, Datta PK, et al: TRIP13 promotes metastasis of colorectal cancer regardless of p53 and microsatellite instability status. Mol Oncol. 14:3007–3029. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Banerjee R, Russo N, Liu M, Basrur V, Bellile E, Palanisamy N, Scanlon CS, van Tubergen E, Inglehart RC, Metwally T, et al: TRIP13 promotes error-prone nonhomologous end joining and induces chemoresistance in head and neck cancer. Nat Commun. 5:45272014. View Article : Google Scholar : PubMed/NCBI

26 

Lan J, Huang J, Tao X, Gao Y, Zhang L, Huang W, Luo J, Liu C, Deng Y, Liu L and Liu X: Evaluation of the TRIP13 level in breast cancer and insights into potential molecular pathways. J Cell Mol Med. 26:2673–2685. 2022. View Article : Google Scholar : PubMed/NCBI

27 

Liu L, Zhang Z, Xia X and Lei J: KIF18B promotes breast cancer cell proliferation, migration and invasion by targeting TRIP13 and activating the Wnt/β-catenin signaling pathway. Oncol Lett. 23:1122022. View Article : Google Scholar : PubMed/NCBI

28 

Li ZH, Lei L, Fei LR, Huang WJ, Zheng YW, Yang MQ, Wang Z, Liu CC and Xu HT: TRIP13 promotes the proliferation and invasion of lung cancer cells via the Wnt signaling pathway and epithelial-mesenchymal transition. J Mol Histol. 52:11–20. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Cai W, Ni W, Jin Y and Li Y: TRIP13 promotes lung cancer cell growth and metastasis through AKT/mTORC1/c-Myc signaling. Cancer Biomark. 30:237–248. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Zhang Q, Dong Y, Hao S, Tong Y, Luo Q and Aerxiding P: The oncogenic role of TRIP13 in regulating proliferation, invasion, and cell cycle checkpoint in NSCLC cells. Int J Clin Exp Pathol. 12:3357–3366. 2019.PubMed/NCBI

31 

Yao J, Zhang X, Li J, Zhao D, Gao B, Zhou H, Gao S and Zhang L: Silencing TRIP13 inhibits cell growth and metastasis of hepatocellular carcinoma by activating of TGF-β1/smad3. Cancer Cell Int. 18:2082018. View Article : Google Scholar : PubMed/NCBI

32 

Garcia MR, Meissburger B, Chan J, de Guia RM, Mattijssen F, Roessler S, Birkenfeld AL, Raschzok N, Riols F, Tokarz J, et al: Trip13 depletion in liver cancer induces a lipogenic response contributing to plin2-dependent mitotic cell death. Adv Sci (Weinh). 9:e21042912022. View Article : Google Scholar : PubMed/NCBI

33 

Dong L, Ding H, Li Y, Xue D, Li Z, Liu Y, Zhang T, Zhou J and Wang P: TRIP13 is a predictor for poor prognosis and regulates cell proliferation, migration and invasion in prostate cancer. Int J Biol Macromol. 121:200–206. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Zeng L, Liu YM, Yang N, Zhang T and Xie H: Hsa_circRNA_100146 promotes prostate cancer progression by upregulating TRIP13 via sponging miR-615-5p. Front Mol Biosci. 8:6934772021. View Article : Google Scholar : PubMed/NCBI

35 

Wang Y, Huang J, Li B, Xue H, Tricot G, Hu L, Xu Z, Sun X, Chang S, Gao L, et al: A small-molecule inhibitor targeting TRIP13 suppresses multiple myeloma progression. Cancer Res. 80:536–548. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Gao Y, Liu S, Guo Q, Zhang S, Zhao Y, Wang H, Li T, Gong Y, Wang Y, Zhang T, et al: Increased expression of TRIP13 drives the tumorigenesis of bladder cancer in association with the EGFR signaling pathway. Int J Biol Sci. 15:1488–1499. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Mohammed aI, Ali ME-H, Mohamed FEA and Abd-Elrehim DM: Immunohistochemical expression of TRIP13 in transitional and squamous cell carcinoma of urinary bladder carcinoma. Minia J Med Res. 2023. View Article : Google Scholar

38 

Zhou KS, Zhang Q, Zhang WT, Liu YY, Wu SS, Zhou J, Wei XD and Song YP: Study on the expression of TRIP13 mRNA in chronic lymphocytic leukemia B lymphocyte and the molecular mechanism of TRIP13 mediated JVM-2 cell proliferation and apoptosis. Zhonghua Xue Ye Xue Za Zhi. 38:618–622. 2017.(In Chinese). PubMed/NCBI

39 

Li W, Zhang G, Li X, Wang X, Li Q, Hong L, Shen Y, Zhao C, Gong X, Chen Y and Zhou J: Thyroid hormone receptor interactor 13 (TRIP13) overexpression associated with tumor progression and poor prognosis in lung adenocarcinoma. Biochem Biophys Res Commun. 499:416–424. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Ju L, Li X, Shao J, Lu R, Wang Y and Bian Z: Upregulation of thyroid hormone receptor interactor 13 is associated with human hepatocellular carcinoma. Oncol Rep. 40:3794–3802. 2018.PubMed/NCBI

41 

Lu W, Mengxuan Z, Ming R, Zixu G, Yong Z, Simin Z, Yang Y, Leqi Q, Kangjie S, Yanlin L, et al: TRIP13/FLNA complex promotes tumor progression and is associated with unfavorable outcomes in melanoma. J Oncol. 2022:14191792022. View Article : Google Scholar : PubMed/NCBI

42 

Tao Y, Yang G, Yang H, Song D, Hu L, Xie B, Wang H, Gao L, Gao M, Xu H, et al: TRIP13 impairs mitotic checkpoint surveillance and is associated with poor prognosis in multiple myeloma. Oncotarget. 8:26718–26731. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Zhang LT, Ke LX, Wu XY, Tian HT, Deng HZ, Xu LY, Li EM and Long L: TRIP13 induces nedaplatin resistance in esophageal squamous cell carcinoma by enhancing repair of DNA damage and inhibiting apoptosis. Biomed Res Int. 2022:72954582022.PubMed/NCBI

44 

Xu H, Ma Z, Mo X, Chen X, Xu F, Wu F, Chen H, Zhou G, Xia H and Zhang C: Inducing synergistic DNA damage by TRIP13 and PARP1 inhibitors provides a potential treatment for hepatocellular carcinoma. J Cancer. 13:2226–2237. 2022. View Article : Google Scholar : PubMed/NCBI

45 

Ye Q, Kim DH, Dereli I, Rosenberg SC, Hagemann G, Herzog F, Tóth A, Cleveland DW and Corbett KD: The AAA+ ATPase TRIP13 remodels HORMA domains through N-terminal engagement and unfolding. EMBO J. 36:2419–2434. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Mapelli M, Massimiliano L, Santaguida S and Musacchio A: The Mad2 conformational dimer: Structure and implications for the spindle assembly checkpoint. Cell. 131:730–743. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Marks DH, Thomas R, Chin Y, Shah R, Khoo C and Benezra R: Mad2 overexpression uncovers a critical role for TRIP13 in mitotic exit. Cell Rep. 19:1832–1845. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Yang M, Li B, Tomchick DR, Machius M, Rizo J, Yu H and Luo X: p31comet blocks Mad2 activation through structural mimicry. Cell. 131:744–755. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Rosenberg SC and Corbett KD: The multifaceted roles of the HORMA domain in cellular signaling. J Cell Biol. 211:745–755. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Zhou K, Zhang W, Zhang Q, Gui R, Zhao H, Chai X, Li Y, Wei X and Song Y: Loss of thyroid hormone receptor interactor 13 inhibits cell proliferation and survival in human chronic lymphocytic leukemia. Oncotarget. 8:25469–25481. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Furlong F, Fitzpatrick P, O'Toole S, Phelan S, McGrogan B, Maguire A, O'Grady A, Gallagher M, Prencipe M, McGoldrick A, et al: Low MAD2 expression levels associate with reduced progression-free survival in patients with high-grade serous epithelial ovarian cancer. J Pathol. 226:746–755. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Zhou XY and Shu XM: TRIP13 promotes proliferation and invasion of epithelial ovarian cancer cells through Notch signaling pathway. Eur Rev Med Pharmacol Sci. 23:522–529. 2019.PubMed/NCBI

53 

Amawi H, Sim HM, Tiwari AK, Ambudkar SV and Shukla S: ABC transporter-mediated multidrug-resistant cancer. Adv Exp Med Biol. 1141:549–580. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Khatami M: Cancer; an induced disease of twentieth century! Induction of tolerance, increased entropy and ‘Dark Energy’: Loss of biorhythms (Anabolism v. Catabolism). Clin Transl Med. 7:202018. View Article : Google Scholar : PubMed/NCBI

55 

Wang DC, Wang W, Zhu B and Wang X: Lung cancer heterogeneity and new strategies for drug therapy. Annu Rev Pharmacol Toxicol. 58:531–546. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Bukowski K, Kciuk M and Kontek R: Mechanisms of multidrug resistance in cancer chemotherapy. Int J Mol Sci. 21:32332020. View Article : Google Scholar : PubMed/NCBI

57 

Vasan N, Baselga J and Hyman DM: A view on drug resistance in cancer. Nature. 575:299–309. 2019. View Article : Google Scholar : PubMed/NCBI

58 

Lu S, Guo M, Fan Z, Chen Y, Shi X, Gu C and Yang Y: Elevated TRIP13 drives cell proliferation and drug resistance in bladder cancer. Am J Transl Res. 11:4397–4410. 2019.PubMed/NCBI

59 

Li C, Xia J, Franqui-Machin R, Chen F, He Y, Ashby TC, Teng F, Xu H, Liu D, Gai D, et al: TRIP13 modulates protein deubiquitination and accelerates tumor development and progression of B cell malignancies. J Clin Invest. 131:e1468932021. View Article : Google Scholar : PubMed/NCBI

60 

Ozols RF and Young RC: High-dose cisplatin therapy in ovarian cancer. Semin Oncol. 12:21–30. 1985.PubMed/NCBI

61 

Markman M: Intraperitoneal cisplatin and carboplatin in the management of ovarian cancer. Semin Oncol. 21:17–19; quiz 20. 581994.PubMed/NCBI

62 

Zoń A and Bednarek I: Cisplatin in ovarian cancer treatment-known limitations in therapy force new solutions. Int J Mol Sci. 24:75852023. View Article : Google Scholar : PubMed/NCBI

63 

Mittica G, Ghisoni E, Giannone G, Genta S, Aglietta M, Sapino A and Valabrega G: PARP inhibitors in ovarian cancer. Recent Pat Anticancer Drug Discov. 13:392–410. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Smith M and Pothuri B: Appropriate selection of PARP inhibitors in ovarian cancer. Curr Treat Options Oncol. 23:887–903. 2022. View Article : Google Scholar : PubMed/NCBI

65 

Jiang X, Li X, Li W, Bai H and Zhang Z: PARP inhibitors in ovarian cancer: Sensitivity prediction and resistance mechanisms. J Cell Mol Med. 23:2303–2313. 2019. View Article : Google Scholar : PubMed/NCBI

66 

Musacchio A and Salmon ED: The spindle-assembly checkpoint in space and time. Nat Rev Mol Cell Biol. 8:379–393. 2007. View Article : Google Scholar : PubMed/NCBI

67 

Lara-Gonzalez P, Westhorpe FG and Taylor SS: The spindle assembly checkpoint. Curr Biol. 22:R966–R980. 2012. View Article : Google Scholar : PubMed/NCBI

68 

Chao WC, Kulkarni K, Zhang Z, Kong EH and Barford D: Structure of the mitotic checkpoint complex. Nature. 484:208–213. 2012. View Article : Google Scholar : PubMed/NCBI

69 

de Cárcer G and Malumbres M: A centrosomal route for cancer genome instability. Nat Cell Biol. 16:504–506. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Sotillo R, Schvartzman JM, Socci ND and Benezra R: Mad2-induced chromosome instability leads to lung tumour relapse after oncogene withdrawal. Nature. 464:436–440. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Bargiela-Iparraguirre J, Prado-Marchal L, Pajuelo-Lozano N, Jiménez B, Perona R and Sánchez-Pérez I: Mad2 and BubR1 modulates tumourigenesis and paclitaxel response in MKN45 gastric cancer cells. Cell Cycle. 13:3590–3601. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Tusell L, Pampalona J, Soler D, Frías C and Genescà A: Different outcomes of telomere-dependent anaphase bridges. Biochem Soc Trans. 38:1698–1703. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Stewénius Y, Gorunova L, Jonson T, Larsson N, Höglund M, Mandahl N, Mertens F, Mitelman F and Gisselsson D: Structural and numerical chromosome changes in colon cancer develop through telomere-mediated anaphase bridges, not through mitotic multipolarity. Proc Natl Acad Sci USA. 102:5541–5546. 2005. View Article : Google Scholar : PubMed/NCBI

74 

Bailey SM and Murnane JP: Telomeres, chromosome instability and cancer. Nucleic Acids Res. 34:2408–2417. 2006. View Article : Google Scholar : PubMed/NCBI

75 

Mills KD, Ferguson DO and Alt FW: The role of DNA breaks in genomic instability and tumorigenesis. Immunol Rev. 194:77–95. 2003. View Article : Google Scholar : PubMed/NCBI

76 

Zhou W, Yang Y, Xia J, Wang H, Salama ME, Xiong W, Xu H, Shetty S, Chen T, Zeng Z, et al: NEK2 induces drug resistance mainly through activation of efflux drug pumps and is associated with poor prognosis in myeloma and other cancers. Cancer Cell. 23:48–62. 2013. View Article : Google Scholar : PubMed/NCBI

77 

Carter SL, Eklund AC, Kohane IS, Harris LN and Szallasi Z: A signature of chromosomal instability inferred from gene expression profiles predicts clinical outcome in multiple human cancers. Nat Genet. 38:1043–1048. 2006. View Article : Google Scholar : PubMed/NCBI

78 

Clairmont CS, Sarangi P, Ponnienselvan K, Galli LD, Csete I, Moreau L, Adelmant G, Chowdhury D, Marto JA and D'Andrea AD: TRIP13 regulates DNA repair pathway choice through REV7 conformational change. Nat Cell Biol. 22:87–96. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Sudakin V, Chan GK and Yen TJ: Checkpoint inhibition of the APC/C in HeLa cells is mediated by a complex of BUBR1, BUB3, CDC20, and MAD2. J Cell Biol. 154:925–936. 2001. View Article : Google Scholar : PubMed/NCBI

80 

Overlack K, Bange T, Weissmann F, Faesen AC, Maffini S, Primorac I, Müller F, Peters JM and Musacchio A: BubR1 promotes Bub3-dependent APC/C inhibition during spindle assembly checkpoint signaling. Curr Biol. 27:2915–2927.e2917. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Burton JL and Solomon MJ: Mad3p, a pseudosubstrate inhibitor of APCCdc20 in the spindle assembly checkpoint. Genes Dev. 21:655–667. 2007. View Article : Google Scholar : PubMed/NCBI

82 

McGranahan N, Burrell RA, Endesfelder D, Novelli MR and Swanton C: Cancer chromosomal instability: Therapeutic and diagnostic challenges. EMBO Rep. 13:528–538. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Lischetti T and Nilsson J: Regulation of mitotic progression by the spindle assembly checkpoint. Mol Cell Oncol. 2:e9704842015. View Article : Google Scholar : PubMed/NCBI

84 

Sudo T, Nitta M, Saya H and Ueno NT: Dependence of paclitaxel sensitivity on a functional spindle assembly checkpoint. Cancer Res. 64:2502–2508. 2004. View Article : Google Scholar : PubMed/NCBI

85 

Wang M, Chen S and Ao D: Targeting DNA repair pathway in cancer: Mechanisms and clinical application. MedComm. 2020.2:654–691. 2021. View Article : Google Scholar : PubMed/NCBI

86 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI

87 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

88 

Deng S, Vlatkovic T, Li M, Zhan T, Veldwijk MR and Herskind C: Targeting the DNA damage response and DNA repair pathways to enhance radiosensitivity in colorectal cancer. Cancers (Basel). 14:48742022. View Article : Google Scholar : PubMed/NCBI

89 

Mirza MR, Pignata S and Ledermann JA: Latest clinical evidence and further development of PARP inhibitors in ovarian cancer. Ann Oncol. 29:1366–1376. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Schettini F, Giudici F, Bernocchi O, Sirico M, Corona SP, Giuliano M, Locci M, Paris I, Scambia G, De Placido S, et al: Poly (ADP-ribose) polymerase inhibitors in solid tumours: Systematic review and meta-analysis. Eur J Cancer. 149:134–152. 2021. View Article : Google Scholar : PubMed/NCBI

91 

Tutt ANJ, Garber JE, Kaufman B, Viale G, Fumagalli D, Rastogi P, Gelber RD, de Azambuja E, Fielding A, Balmaña J, et al: Adjuvant olaparib for patients with BRCA1- or BRCA2-mutated breast cancer. N Engl J Med. 384:2394–2405. 2021. View Article : Google Scholar : PubMed/NCBI

92 

Carreira S, Porta N, Arce-Gallego S, Seed G, Llop-Guevara A, Bianchini D, Rescigno P, Paschalis A, Bertan C, Baker C, et al: Biomarkers associating with PARP inhibitor benefit in prostate cancer in the TOPARP-B trial. Cancer Discov. 11:2812–2827. 2021. View Article : Google Scholar : PubMed/NCBI

93 

Mateo J, Porta N, Bianchini D, McGovern U, Elliott T, Jones R, Syndikus I, Ralph C, Jain S, Varughese M, et al: Olaparib in patients with metastatic castration-resistant prostate cancer with DNA repair gene aberrations (TOPARP-B): A multicentre, open-label, randomised, phase 2 trial. Lancet Oncol. 21:162–174. 2020. View Article : Google Scholar : PubMed/NCBI

94 

Maughan BL and Antonarakis ES: Olaparib and rucaparib for the treatment of DNA repair-deficient metastatic castration-resistant prostate cancer. Expert Opin Pharmacother. 22:1625–1632. 2021. View Article : Google Scholar : PubMed/NCBI

95 

Cleary JM, Wolpin BM, Dougan SK, Raghavan S, Singh H, Huffman B, Sethi NS, Nowak JA, Shapiro GI, Aguirre AJ and D'Andrea AD: Opportunities for utilization of DNA repair inhibitors in homologous recombination repair-deficient and proficient pancreatic adenocarcinoma. Clin Cancer Res. 27:6622–6637. 2021. View Article : Google Scholar : PubMed/NCBI

96 

van Waardenburg R and Yang ES: Targeting DNA repair pathways to overcome cancer drug resistance. Cancer Drug Resist. 4:837–841. 2021.PubMed/NCBI

97 

Ghosh S, Mazumdar T, Xu W, Powell RT, Stephan C, Shen L, Shah PA, Pickering CR, Myers JN, Wang J, et al: Combined TRIP13 and aurora kinase inhibition induces apoptosis in human papillomavirus–driven cancers. Clin Cancer Res. 28:4479–4493. 2022. View Article : Google Scholar : PubMed/NCBI

98 

Chang H and Zou Z: Targeting autophagy to overcome drug resistance: Further developments. J Hematol Oncol. 13:1592020. View Article : Google Scholar : PubMed/NCBI

99 

Ahmadi-Dehlaghi F, Mohammadi P, Valipour E, Pournaghi P, Kiani S and Mansouri K: Autophagy: A challengeable paradox in cancer treatment. Cancer Med. 12:11542–11569. 2023. View Article : Google Scholar : PubMed/NCBI

100 

Salimi-Jeda A, Ghabeshi S, Pour ZGM, Jazaeri EO, Araiinejad M, Sheikholeslami F, Abdoli M, Edalat M and Abdoli A: Autophagy modulation and cancer combination therapy: A smart approach in cancer therapy. Cancer Treat Res Commun. 30:1005122022. View Article : Google Scholar : PubMed/NCBI

101 

Levine B and Kroemer G: Biological functions of autophagy genes: A disease perspective. Cell. 176:11–42. 2019. View Article : Google Scholar : PubMed/NCBI

102 

Galluzzi L and Green DR: Autophagy-independent functions of the autophagy machinery. Cell. 177:1682–1699. 2019. View Article : Google Scholar : PubMed/NCBI

103 

Dikic I and Elazar Z: Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol. 19:349–364. 2018. View Article : Google Scholar : PubMed/NCBI

104 

Wu M and Zhang P: EGFR-mediated autophagy in tumourigenesis and therapeutic resistance. Cancer Lett. 469:207–216. 2020. View Article : Google Scholar : PubMed/NCBI

105 

Li YJ, Lei YH, Yao N, Wang CR, Hu N, Ye WC, Zhang DM and Chen ZS: Autophagy and multidrug resistance in cancer. Chin J Cancer. 36:522017. View Article : Google Scholar : PubMed/NCBI

106 

Amaravadi RK, Kimmelman AC and Debnath J: Targeting autophagy in cancer: Recent advances and future directions. Cancer Discov. 9:1167–1181. 2019. View Article : Google Scholar : PubMed/NCBI

107 

Xiao Z, Li M, Zhang X, Rong X and Xu H: TRIP13 overexpression promotes gefitinib resistance in non-small cell lung cancer via regulating autophagy and phosphorylation of the EGFR signaling pathway. Oncol Rep. 49:842023. View Article : Google Scholar : PubMed/NCBI

108 

Sharma P, Hu-Lieskovan S, Wargo JA and Ribas A: Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 168:707–723. 2017. View Article : Google Scholar : PubMed/NCBI

109 

Agarwal S, Afaq F, Bajpai P, Kim HG, Elkholy A, Behring M, Chandrashekar DS, Diffalha SA, Khushman M, Sugandha SP, et al: DCZ0415, a small-molecule inhibitor targeting TRIP13, inhibits EMT and metastasis via inactivation of the FGFR4/STAT3 axis and the Wnt/β-catenin pathway in colorectal cancer. Mol Oncol. 16:1728–1745. 2022. View Article : Google Scholar : PubMed/NCBI

110 

Qie S and Diehl JA: Cyclin D1, cancer progression, and opportunities in cancer treatment. J Mol Med (Berl). 94:1313–1326. 2016. View Article : Google Scholar : PubMed/NCBI

111 

Lindahl T and Barnes DE: Repair of endogenous DNA damage. Cold Spring Harb Symp Quant Biol. 65:127–133. 2000. View Article : Google Scholar : PubMed/NCBI

112 

Sarangi P, Clairmont CS, Galli LD, Moreau LA and D'Andrea AD: p31(comet) promotes homologous recombination by inactivating REV7 through the TRIP13 ATPase. Proc Natl Acad Sci USA. 117:26795–26803. 2020. View Article : Google Scholar : PubMed/NCBI

113 

Corbett KD: p31comet and TRIP13 recycle Rev7 to regulate DNA repair. Proc Natl Acad Sci USA. 117:27761–27763. 2020. View Article : Google Scholar : PubMed/NCBI

114 

Oser MG, Fonseca R, Chakraborty AA, Brough R, Spektor A, Jennings RB, Flaifel A, Novak JS, Gulati A, Buss E, et al: Cells lacking the RB1 tumor suppressor gene are hyperdependent on aurora B kinase for survival. Cancer Discov. 9:230–247. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Gong X, Du J, Parsons SH, Merzoug FF, Webster Y, Iversen PW, Chio LC, Van Horn RD, Lin X, Blosser W, et al: Aurora A kinase inhibition is synthetic lethal with loss of the RB1 tumor suppressor gene. Cancer Discov. 9:248–263. 2019. View Article : Google Scholar : PubMed/NCBI

116 

Schvartzman JM, Duijf PH, Sotillo R, Coker C and Benezra R: Mad2 is a critical mediator of the chromosome instability observed upon Rb and p53 pathway inhibition. Cancer Cell. 19:701–714. 2011. View Article : Google Scholar : PubMed/NCBI

117 

Li Z, Liu J, Chen T, Sun R, Liu Z, Qiu B, Xu Y and Zhang Z: HMGA1-TRIP13 axis promotes stemness and epithelial mesenchymal transition of perihilar cholangiocarcinoma in a positive feedback loop dependent on c-Myc. J Exp Clin Cancer Res. 40:862021. View Article : Google Scholar : PubMed/NCBI

118 

Zhang X, Zhou J, Xue D, Li Z, Liu Y and Dong L: MiR-515-5p acts as a tumor suppressor via targeting TRIP13 in prostate cancer. Int J Biol Macromol. 129:227–232. 2019. View Article : Google Scholar : PubMed/NCBI

119 

Chen Y, Chen D, Qin Y, Qiu C, Zhou Y, Dai M, Li L, Sun Q and Jiang Y: TRIP13, identified as a hub gene of tumor progression, is the target of microRNA-4693-5p and a potential therapeutic target for colorectal cancer. Cell Death Discov. 8:352022. View Article : Google Scholar : PubMed/NCBI

120 

Zhu MX, Wei CY, Zhang PF, Gao DM, Chen J, Zhao Y, Dong SS and Liu BB: Elevated TRIP13 drives the AKT/mTOR pathway to induce the progression of hepatocellular carcinoma via interacting with ACTN4. J Exp Clin Cancer Res. 38:4092019. View Article : Google Scholar : PubMed/NCBI

121 

Arun G, Diermeier SD and Spector DL: Therapeutic targeting of long non-coding RNAs in cancer. Trends Mol Med. 24:257–277. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2023
Volume 50 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao L, Ye S, Jing S, Gao Y and He T: Targeting TRIP13 for overcoming anticancer drug resistance (Review). Oncol Rep 50: 202, 2023
APA
Zhao, L., Ye, S., Jing, S., Gao, Y., & He, T. (2023). Targeting TRIP13 for overcoming anticancer drug resistance (Review). Oncology Reports, 50, 202. https://doi.org/10.3892/or.2023.8639
MLA
Zhao, L., Ye, S., Jing, S., Gao, Y., He, T."Targeting TRIP13 for overcoming anticancer drug resistance (Review)". Oncology Reports 50.5 (2023): 202.
Chicago
Zhao, L., Ye, S., Jing, S., Gao, Y., He, T."Targeting TRIP13 for overcoming anticancer drug resistance (Review)". Oncology Reports 50, no. 5 (2023): 202. https://doi.org/10.3892/or.2023.8639