Skip to main content
Cancers logoLink to Cancers
. 2025 Aug 26;17(17):2787. doi: 10.3390/cancers17172787

Unlocking the Therapeutic Potential of DNA-PKcs in Cancer: Comprehensive Insights into Mechanisms and Clinical Applications

Tong Zheng 1,2,3, Chao Sun 1,3, Cijun Yun 1,2,3, Hui Wang 1,2,3,4,*, Xiongxiong Liu 1,2,3,4,*
Editors: David Wong, Nicola Curtin
PMCID: PMC12427243  PMID: 40940882

Simple Summary

Cancer remains a major global health challenge. This review focuses on DNA-PKcs, a protein that enables cancer cells to repair DNA damage caused by radiotherapy and chemotherapy, promoting tumor survival. We synthesize recent evidence on how DNA-PKcs drives tumor progression and therapy resistance, highlighting its potential as a therapeutic target. Current advances in DNA-PKcs inhibitors are examined, including their promise in combination therapies and hurdles in clinical development. Uniquely, this review connects DNA-PKcs to emerging roles in immunomodulation and cancer metabolism, evaluates next-generation inhibitors, and proposes solutions to overcome translational challenges.

Keywords: DNA-PKcs, targeted therapy, DNA repair, tumor resistance

Abstract

Cancer remains one of the most pressing global health challenges, with current therapies often hindered by limited efficacy and the emergence of resistance. The DNA-dependent protein kinase catalytic subunit (DNA-PKcs), a key regulator of DNA repair and cell cycle progression, plays a critical role in maintaining genomic stability, and growing evidence indicates its dysregulation in various cancers, with overexpression frequently associated with aggressive tumor phenotypes. To evaluate DNA-PKcs as a therapeutic target, we systematically analyzed literature from PubMed and Web of Science (2000–2024) using keywords including DNA-PKcs, targeted therapy, DNA repair, and tumor resistance following PRISMA guidelines, with 185 of 1250 initial records meeting inclusion criteria after screening. The review explores the multifaceted roles of DNA-PKcs in tumor biology and resistance mechanisms, evaluates the current landscape of DNA-PKcs inhibitors, including their clinical progress and combination strategies with radiotherapy and chemotherapy, and discusses key determinants of therapeutic efficacy, such as tumor type and mutation status. Additionally, it uniquely integrates emerging insights into the roles of DNA-PKcs in immunomodulation and metabolism, critically assesses next-generation inhibitors, and proposes strategies to address remaining challenges. Through this comprehensive analysis, we highlight the therapeutic potential of DNA-PKcs inhibition as a novel strategy to circumvent treatment resistance, providing innovative insights for optimizing cancer management, especially for aggressive tumor subtypes, thereby advancing drug discovery efforts and paving the way for more effective therapeutic interventions in clinical practice.

1. Introduction

Cancer continues to pose a formidable threat to global health, with the World Health Organization (WHO) reporting nearly 10 million cancer-related deaths in 2020 alone. While significant progress has been made in surgical, radiation, and systemic therapies, critical challenges persist, including intrinsic and acquired treatment resistance [1,2], therapy-associated toxicities [3,4,5], and interpatient heterogeneity [6,7]. These limitations highlight the urgent need for novel, targeted therapeutic strategies to enhance treatment efficacy and patient survival [8].

The DNA-dependent protein kinase catalytic subunit (DNA-PKcs), a core component of the DNA-PK complex, plays a pivotal role in repairing DNA double-strand breaks (DSBs)—a critical mechanism underlying cancer cell survival and genomic instability. Notably, cancer cells exhibit heightened DSB repair capacity, enabling uncontrolled proliferation and resistance to genotoxic therapies [9,10]. Beyond its canonical role in DSB repair, DNA-PKcs regulates diverse cellular processes, including cell cycle progression and apoptosis. Importantly, DNA-PKcs is frequently overexpressed or hyperactivated in multiple malignancies, such as nasopharyngeal carcinoma [11], breast cancer [12], and gastric cancer [13], with preclinical studies demonstrating that its inhibition effectively suppresses tumor growth. These findings position DNA-PKcs as a compelling therapeutic target in oncology.

This review aims to critically evaluate the therapeutic potential of DNA-PKcs inhibition in cancer treatment. We will (1) elucidate the biological functions of DNA-PKcs in tumorigenesis and therapy resistance, (2) assess the current landscape of DNA-PKcs inhibitors and their clinical progress, and (3) discuss the rationale for combining DNA-PKcs-targeted therapies with radiotherapy, chemotherapy, and emerging modalities. By synthesizing these insights, we seek to underscore DNA-PKcs as a high-value target and provide a roadmap for future research to optimize cancer therapeutics.

2. The Structure of DNA-PKcs

DNA-PKcs, encoded by the PRKDC gene, is a 460 kDa multifunctional serine/threonine kinase belonging to the phosphatidylinositol 3-kinase-related kinase (PIKK) family, which includes other DNA damage response kinases like ATM and ATR [14,15]. This large protein (4128 amino acids) features an N-terminal HEAT repeat domain that forms an open ring structure essential for DNA-PK complex assembly and non-homologous end joining (NHEJ), with its binding affinity enhanced by a leucine-rich region [16,17,18]. The C-terminal region contains characteristic PIKK family domains, including a catalytic kinase domain that phosphorylates substrates involved in NHEJ, cellular metabolism, and apoptosis regulation [16,19,20]. Key regulatory domains include the FAT and FATC regions that maintain protein stability and facilitate substrate interactions, as well as the FRB domain which mediates mTOR binding for metabolic regulation [16,19,20] (Figure 1). Importantly, structural studies have identified conserved phosphorylation clusters (JK, ABCDE, and PQR) that critically regulate DNA-PKcs activation, repair complex assembly, and NHEJ efficiency [19,20,21,22], highlighting the sophisticated molecular architecture that enables its diverse cellular functions.

Figure 1.

Figure 1

Domain architecture and functional phosphorylation sites of DNA-PKcs. Schematic representation of the major structural domains of DNA-PKcs, including the N-terminal HEAT repeats, FAT (FRAP-ATM-TRRAP), FRB (FKBP12-rapamycin binding), kinase, and C-terminal FATC domains. Key phosphorylation clusters (JK, PQR, and ABCDE) that regulate DNA-PKcs activity and function in DNA damage response. Phosphorylation at these sites modulates DNA-PKcs activation, autophosphorylation, and interaction with repair machinery.

3. The Function of DNA-PKcs in Tumor Biology

3.1. The Classical Function of DNA-PKcs

DNA-PKcs plays pivotal roles in multiple DNA repair pathways, with its most well-characterized function in non-homologous end joining (NHEJ). Upon DNA double-strand break (DSB) formation, DNA-PKcs is recruited to damage sites where it complexes with Ku70/Ku80 to form DNA-PK [23,24,25]. This complex initiates NHEJ by recognizing broken DNA ends and facilitating their repair through autophosphorylation and substrate phosphorylation [26,27,28,29,30,31].

Beyond NHEJ, DNA-PKcs is essential for V(D)J recombination, the process that generates antibody diversity in developing lymphocytes [32,33]. It mediates DSB recognition and processing during immunoglobulin gene rearrangement while regulating key factors including Artemis and XRCC4 through its kinase activity [34,35,36].

DNA-PKcs also influences the choice between NHEJ and homologous recombination (HR) repair pathways. Through competitive phosphorylation with other PIKK family members, it modulates HR proteins such as BRCA1 and EXO1 [1,37,38,39,40]. While its role in single-strand break repair remains less understood, emerging evidence suggests single-stranded DNA can activate DNA-PKcs kinase function [41]. These DNA repair functions are particularly crucial for cancer cells, which depend on DNA-PKcs to maintain genomic stability during rapid proliferation and increased DNA damage burden (Figure 2).

Figure 2.

Figure 2

DNA-PKcs plays complex and indispensable roles extending far beyond its canonical functions, including DNA repair, cell cycle regulation, transcriptional control, and immune modulation. In DNA repair, it is recruited to double-strand breaks (DSBs) via the Ku70/Ku80 complex, where phosphorylation at Thr2609/Ser2056 clusters activates non-homologous end joining (NHEJ) while competitively inhibiting homologous recombination (HR) through BRCA1 and EXO1 phosphorylation. For cell cycle regulation, DNA-PKcs attenuates ATM-mediated G1/S arrest by phosphorylating MDC1, ensures proper chromosome segregation by modulating Cyclin B1 stability through the Cdh1-APC/C pathway and phosphorylating Bub3, and stabilizes replication forks via ATR and PIDD to prevent replication stress during S phase. In transcriptional regulation, it enhances RNA polymerase II activity and facilitates transcription factor recruitment. In immune modulation, it is essential for generating antigen receptor diversity and modulating immune factor expression, highlighting its multifaceted roles in cellular homeostasis.

3.2. Cell Cycle Regulation by DNA-PKcs

DNA-PKcs plays a critical role in coordinating cell cycle progression through multiple regulatory mechanisms (Figure 2). As a key DNA damage sensor, it modulates checkpoint responses at both G1/S and G2/M transitions. Through phosphorylation of MDC1, DNA-PKcs attenuates ATM-mediated G1/S checkpoint activation [42,43], while simultaneously regulating Cyclin B1 stability via the Cdh1-APC/C ubiquitination pathway to control G2/M transition [44]. Notably, DNA-PKcs overexpression can trigger Chk2 activation, leading to centrosome amplification and G2/M checkpoint disruption [45].

The kinase exerts broad influence over cell cycle progression through the c-Myc/E2F axis. By maintaining c-Myc stability [46], DNA-PKcs indirectly regulates cyclin D/CDK4/6 expression and subsequent pRb-E2F signaling [47,48,49]. Reciprocal regulation occurs through E2F binding sites in DNA-PKcs promoter regions [50], suggesting a feedback mechanism. During S phase, DNA-PKcs phosphorylates critical substrates including PRA2 and BRCA1 [38,51], while participating in Chk1-Claspin complex formation to maintain genomic stability [52]. Its functional interplay with other PIKK members is dose-dependent, with low radiation activating ATM and higher doses engaging both ATM and ATR for G2/M checkpoint control [53].

DNA-PKcs also safeguards chromosomal integrity throughout the cell cycle. It collaborates with PIDD to stabilize replication forks through ATR-mediated phosphorylation [54,55], while suppressing aberrant chromosome rearrangements via DNA end protection [56]. During mitosis, DNA-PKcs contributes to accurate chromosome segregation through Bub3 phosphorylation [57] and maintains telomere stability via interactions with TRF2 [58,59] and hnRNP-A1 phosphorylation [60].

While these findings establish DNA-PKcs as a master regulator of cell cycle progression, the complete mechanistic remains to be elucidated. Further investigation of these pathways may yield novel therapeutic strategies for cancer treatment by targeting DNA-PKcs-mediated cell cycle control.

3.3. DNA-PKcs in Tumor Immunogenicity

The emerging role of DNA-PKcs in immunoregulation presents new opportunities for cancer immunotherapy (Figure 3). As a key mediator of V(D)J recombination, DNA-PKcs deficiency results in severe immunodeficiency, characterized by reduced immune cell populations and radiosensitivity in murine models [61,62,63,64]. Mechanistically, DNA-PKcs regulates T cell function through multiple pathways: (1) modulating Gata3-mediated cytokine production (IL-4, IL-5, IL-13) in CD4+ T cells [65]; (2) activating ZAK/mTOR signaling to maintain senescent T cell proliferation [66]; and (3) phosphorylating CHK2 and Egr1 (Early growth response protein 1) to upregulate IL-2 expression [67,68].

Figure 3.

Figure 3

Role of DNA-PKcs in Tumor Immunogenicity. It modulates immune responses through: (1) Transcriptional regulation of immune-related factors (2) Phosphorylation-mediated inhibition of cGAS, thereby suppressing the cGAS/STING innate immune pathway (counteracted by ARIH1 through proteasomal degradation of DNA-PKcs) (3) RNF144A-dependent regulation of PD-L1 expression, potentially limiting immune evasion in cancer and (4) In B cells, DNA-PKcs is essential for immunoglobulin class switch recombination (CSR), and its deficiency leads to genomic instability. In T cells, DNA-PKcs modulates Gata3-dependent cytokine production, sustains proliferative capacity by activating the ZAK/mTOR signaling axis, phosphorylates CHK2 and Egr1, thereby enhancing IL-2 expression and promoting T cell activation.

In B cell immunity, DNA-PKcs is indispensable for immunoglobulin class switch recombination (CSR). DNA-PKcs-deficient B cells exhibit defective CSR with increased alternative end joining (aEJ) activity, leading to genomic instability through non-productive translocations and deletions [69,70,71,72]. Phosphorylation at Thr2609 appears critical for maintaining canonical NHEJ during CSR [72], suggesting therapeutic potential for modulating DNA-PKcs activity.

The kinase also interfaces with innate immunity through the cGAS-STING pathway [73,74]. While DNA-PKcs phosphorylates and inhibits cGAS [75,76], its degradation by ARIH1 enhances STING-dependent immune activation [76]. This regulatory interplay extends to other immune pathways (NF-κB, JNK, TNF-α) [77,78,79,80], though context-dependent effects are observed—dietary restriction activates DNA-PKcs/p53-mediated tumor suppression [81,82,83], highlighting its pleiotropic functions.

Notably, DNA-PKcs interacts with immune checkpoint regulation [84,85]. It maintains PD-L1 expression [86,87,88], while the E3 ligase RNF144A coordinately degrades both DNA-PKcs and PD-L1 [87,89]. These findings position DNA-PKcs as a promising target for combination immunotherapy, though its complex immunomodulatory roles necessitate careful therapeutic development.

3.4. Transcriptional Regulation by DNA-PKcs

DNA-PKcs plays a pivotal role in transcriptional regulation through multiple mechanisms (Figure 2). Initial studies in vivo experiments show that DNA-PKcs is essential for p53-dependent high-level transcription [90]. This kinase regulates RNA polymerase II (RNAPII) activity by facilitating its phosphorylation and formation of transcription factor complexes [91,92], while also being recruited by hypoxia-inducible factor-1 to reactivate transcription under hypoxic conditions [93,94,95]. Additionally, DNA-PKcs participates in ribosomal biogenesis through phosphorylation of hnRNP and pre-rRNA [96,97], and modulates the activity of various transcription factors including: (1) Core transcriptional regulators (TBP, USF, FoxA2) [98,99,100]; (2) Metabolic/survival mediators (NF-κB, Myc, Sp1) [101,102,103]; (3) Immune response controllers (Egr1, NFAT) [68].

Dysregulation of these DNA-PKcs-mediated processes contributes to multiple pathologies such as tumorigenesis, renal, heart and neurological diseases [104,105,106]. AR variants (AR-Vs), which are unconventional nuclear hormone receptor family transcription factors, have their transcriptional activity enhanced by DNA-PKcs binding to the transcriptional elements and DNA-PKcs also participates in the mRNA processing of AR-Vs together with RNA-binding protein RBMX, eventually leading to increased expression of AR-Vs in advanced prostate cancer and resistance to traditional AR-targeted therapy [107].

3.5. Elevated DNA-PKcs Levels as a Driver of Tumor Progression

Elevated DNA-PKcs expression is strongly correlated with enhanced metastatic potential and poor clinical prognosis across multiple cancer types, functioning through both direct and indirect oncogenic mechanisms (Figure 4). At the direct level, DNA-PKcs facilitates cancer cell survival by promoting efficient repair of DNA double-strand breaks (DSBs) through increased protein expression and/or enhanced kinase activity. Indirectly, it drives tumor progression by orchestrating metabolic reprogramming, modulating gene expression networks, and reshaping the tumor microenvironment [1,4,62].

Figure 4.

Figure 4

DNA-PKcs upregulation drives multifaceted oncogenic pathways in cancer progression. DNA-PKcs forms a complex with Ku70/Ku80 at double-strand breaks (DSBs), where it phosphorylates Thr2609/Ser2056 clusters to activate NHEJ repair. Through the TGF-β/Smad3 and PI3K/Akt/mTOR pathways, DNA-PKcs triggers epithelial–mesenchymal transition (EMT). It also phosphorylates Sp1, which promotes PRKDC mRNA expression. As non-coding RNA effects on PRKDC mRNA decrease, DNA-PKcs levels rise further, forming a positive feedback loop. This process activates glycolysis through enzymatic actions, supplying energy to cancer cells. Additionally, DNA-PKcs promotes vascular endothelial growth factor expression, supporting angiogenesis. High DNA-PKcs levels are also associated with PD-L1 overexpression, the combination of DNA-PKcs inhibitors with immunotherapy represents a promising therapeutic strategy.

The transcriptional and post-transcriptional regulation of DNA-PKcs plays a pivotal role in cancer progression, as exemplified by its interaction with transcription factor Sp1 in hepatocellular carcinoma, colorectal cancer, and gliomas. This interaction not only enhances PRKDC promoter binding and transcription but may also establish a self-reinforcing loop through Sp1 phosphorylation by DNA-PKcs, thereby promoting tumor aggressiveness and therapy resistance [12,102,108,109,110,111]. Similar regulatory mechanisms have been observed in cervical cancer [112]. Non-coding RNAs further contribute to this complex regulatory network, with miR-101 downregulation increasing PRKDC expression [113], while ScaRNA2 and linc00312 inhibit DNA repair by binding DNA-PKcs [114,115]. The lncRNA LINP1 facilitates DNA-PK complex formation during NHEJ [116], and circ_PRDKC modulates drug resistance and cell cycle checkpoints through miRNA sponging [117,118]. DNA-PKcs also forms critical interactions with other molecules, such as the DNA-PKcs/Survivin heterotetramer that enhances DSB repair while suppressing apoptosis [119,120], and its autophosphorylation at Thr2609 and Ser2056 regulates DSB repair efficiency and mitotic progression, though the full implications of these modifications remain incompletely understood [22,72,121]. The regulatory complexity is further compounded by phosphorylation events mediated by other kinases, particularly members of the PIKK family [122].

Recent advances have illuminated DNA-PKcs’ role in metabolic reprogramming and transcriptional control across various cancers. In castration-resistant prostate cancer (CRPC), it upregulates glycolysis through hexokinase 2 and pyruvate kinase M2 activation [123] while simultaneously promoting androgen receptor (AR) transcription [107]. Small cell lung cancer (SCLC) studies reveal its cooperation with OCT4 to activate Myc [101], and in pancreatic cancer, it modulates survival through the PI3K/Akt/mTOR pathway [46,110]. DNA-PKcs-mediated Smad3 phosphorylation enhances TGF-β1-induced epithelial–mesenchymal transition (EMT), contributing to metastasis in colorectal, gastric, and liver cancers [124,125,126,127]. Notably, in gliomas, the aberrantly expressed TRIM24 recruits DNA-PKcs and PHAX, leading to TRIM24 phosphorylation and subsequent oncogenic transcriptome alterations [128].

Although research on the role of DNA-PKcs in the tumor microenvironment (TME) remains limited, several significant findings have emerged that warrant attention. Studies demonstrate that DNA-PKcs facilitates Drp1 phosphorylation, a key mitochondrial fission protein, thereby accelerating mitochondrial fragmentation and modulating angiotensin II (Ang II)-induced vascular remodeling [129]. In TPEN-treated colon cancer cells, DNA-PKcs cooperates with Chk1 to mediate ROS-dependent DNA damage [130], revealing the therapeutic potential of heavy metal chelators in oncology. Notably, metastatic triple-negative breast cancer (TNBC) often develops immunotherapy resistance due to compromised immune cell infiltration and impaired inflammatory responses within the TME [131]. Intriguingly, DNA-PKcs inhibition has been shown to enhance immune cell infiltration and upregulate immunomodulatory factors, thereby reshaping the TME and potentiating anti-tumor immunity in TNBC [132]. Another mechanistic study in gastric cancer identified that hsa_circ_0136666 competitively binds miR-375-3p to elevate DNA-PKcs expression, consequently inducing PD-L1 overexpression and facilitating immune evasion88. Growing evidence further implicates DNA-PKcs in regulating oxidative stress responses, Akt signaling, and Smad/TGF-β1 pathways, which collectively influence tumor invasion and metastatic potential [91,92,124,127,133,134].

Emerging clinical observations suggest an unexpected association between DNA-PKcs downregulation and gastric cancer progression. A comprehensive clinicopathological analysis revealed that gastric cancer patients lacking DNA-PKcs expression exhibited significantly worse survival outcomes compared to those with preserved expression (p = 0.004) [135]. Moreover, DNA-PKcs loss showed strong correlation with advanced disease stage (p < 0.001) [135], implying potential tumor-suppressive functions that merit further investigation. While most therapeutic strategies targeting DNA-PKcs overexpression have shown encouraging clinical efficacy [136,137,138,139,140], the protein’s pleiotropic functions underscore the need for more sophisticated research approaches. Future investigations employing single-cell omics and spatial omics technologies will be crucial for elucidating the complex biology of DNA-PKcs and optimizing targeted therapeutic interventions.

4. Therapeutic Strategies Targeting DNA-PKcs

Given the critical role of DNA-PKcs in cancer cell proliferation and treatment resistance, considerable efforts have been devoted to developing DNA-PKcs inhibitors, many of which are now under preclinical and clinical evaluation. Targeting DNA-PKcs offers a promising strategy to overcome therapeutic resistance—a major challenge in oncology that frequently leads to tumor relapse and disease progression.

Early-generation inhibitors such as NU7026 and NU7441 attracted substantial interest due to their high selectivity for DNA-PKcs inhibition, yet they remain confined to preclinical studies [141,142]. In recent years, however, a new wave of next-generation DNA-PKcs inhibitors has entered clinical trials, either as standalone therapies or in combination with radiotherapy and chemotherapy. The following section highlights recent advances in DNA-PKcs-targeted therapies, focusing on their synergistic effects with conventional treatments and key factors influencing therapeutic efficacy (Table 1).

Table 1.

Clinical Trials Focusing on DNA-PKcs.

DNA-PKcs Inhibitos Target Combination Therapy Clinical Trial Phase Types of Cancer Treatment Clinical Trial Registration
Peposertib DNA-PKcs Monotherapy I Advanced Solid Tumors NCT02516813
Radiotherapy I Glioblastoma, Gliosarcoma NCT04555577
Monotherapy I/II Locally Advanced Rectal Cancer NCT03770689
Radiotherapy, Avelumab I/II Cholangiocarcinoma, Gallbladder Carcinoma, Stage III Gallbladder Cancer AJCC v8, and 5 more NCT04068194
Radium-223 Dichloride, Avelumab I/II Metastatic Castration-Resistant Prostate Carcinoma, Metastatic Malignant Neoplasm in the Bone, Metastatic Malignant Neoplasm in the Lymph Nodes, and 1 more NCT04071236
Lutetium 177 Dotatate I Neuroendocrine Neoplasm NCT04750954
CC-115 DNA-PKcs & mTOR Monotherapy I Glioblastoma Multiforme, Glioblastoma Multiforme, Prostate Cancer, and 3 more NCT01353625
Temozolomide, Neratinib II Glioblastoma NCT02977780
Enzalutamide I Prostate Cancer, Castration Resistant Prostate Cancer NCT02833883
XRD-0394 DNA-PKcs & ATM Radiation: Palliative radiotherapy I Metastasis, Locally Advanced Solid Tumor, Recurrent Cancer NCT05002140
AZD7648 DNA-PKcs Radiotherapy I Soft Tissue Sarcoma Adult NCT05116254
Pegylated Liposomal Doxorubicin I/II Advanced Malignancies NCT03907969
M9831 DNA-PKcs Pegylated Liposomal Doxorubicin I Advanced Solid Tumor NCT02644278
AsiDNA DNA-PKcs Monotherapy I Advanced Cancer NCT03579628
Niraparib, Olaparib I/II Ovarian Cancer NCT04826198
Monotherapy I/II Recurrent High-grade Glioma NCT05394558
Olaparib I/II Metastatic Castration-resistant Prostate Cancer, Recurrent Epithelial Ovarian Cancer, Breast Cancer NCT05700669

4.1. Current Landscape of DNA-PKcs Inhibitors

Preclinical studies have identified NU7026 and NU7441 as promising DNA-PKcs inhibitors, yet their clinical translation has been hindered by pharmacokinetic limitations including rapid blood clearance, poor oral bioavailability, and off-target effects on the mTOR/PI3K pathway [143,144]. These challenges have spurred the development of novel, more clinically viable inhibitors.

4.1.1. Peposertib

Peposertib is a highly potent and selective small-molecule inhibitor of DNA-PKcs, currently under advanced clinical evaluation. Preclinical studies in cervical cancer xenograft models demonstrated significant target inhibition following oral administration. In the first-in-human Phase I trial (NCT02316197), 400 mg twice daily (BID) was established as the recommended Phase II dose (RP2D), with dose-limiting toxicities—primarily manageable gastrointestinal events—observed at 300 mg BID. Pharmacodynamic analyses confirmed target engagement, as evidenced by reduced phosphorylated DNA-PK levels in peripheral blood mononuclear cells, supporting its progression into combination therapy trials [145].

4.1.2. CC-115

CC-115 is a promising dual-target inhibitor that selectively targets mTOR and DNA-PKcs, modulating the cell cycle and DSB repair, and it has shown efficacy in renal cancer and NSCLC cell lines [146,147,148]. In the first-in-human Phase I study, CC-115 showed notable efficacy and tolerability in patients with advanced tumors. The half-life and blood concentration were within acceptable ranges, and the RP2D was set at 10 mg BID. Remarkably, a patient with endometrial cancer achieved complete regression of lung and lymph node lesions after 10 cycles, which was maintained for four years. Additionally, the study suggests that CC-115 may be effective against a broader range of solid tumors than previously thought [149]. The Ib phase multi-center trial demonstrated the feasibility of combining 5 mg BID CC-115 with the AR inhibitor enzalutamide in patients with mCRPC. Notably, differences in PSA50 (Prostate-specific Antigen 50% decline from baseline, and the same applies to PSA90) and PSA90 between patients with and without PI3K pathway alterations underscored the importance of the PI3K pathway in this combination therapy [150].

4.1.3. XRD-0394

XRD-0394 is an orally bioavailable dual inhibitor with high potency and specificity against ATM and DNA-PKcs. By disrupting these kinases in cancer cells, XRD-0394 enhances tumor sensitivity to radiotherapy (RT) [151].

4.1.4. AZD7648

AZD7648 has shown promising potential in combination with RT, chemotherapy, and immunotherapy [152,153,154,155]. A completed Phase I/II trial (NCT03907969) evaluated AZD7648 in combination with pegylated liposomal doxorubicin (PLD), while another ongoing trial (NCT05116254) is investigating its synergy with RT.

4.1.5. M9831 (VX-984)

M9831 enhances the radiosensitivity of cancer cells in a concentration-dependent manner, though its efficacy as a monotherapy requires further investigation [137,156]. A Phase I trial (NCT02644278) assessing M9831 in combination with PLD for advanced solid tumors has been completed.

4.1.6. AsiDNA

Unlike conventional DNA-PKcs inhibitors, AsiDNA operates via a unique mechanism: it is a double-stranded DNA decoy that mimics DSBs, sequestering key NHEJ repair proteins, including DNA-PKcs. By depleting available DNA-PKcs, AsiDNA impairs the repair of endogenous DNA damage in cancer cells, ultimately triggering cell death [157,158,159]. This mechanism confers AsiDNA with a distinct advantage in overcoming drug resistance, making it a particularly promising therapeutic agent [157,160].

4.2. Combination Therapies Targeting DNA-PKcs (Table 2)

4.2.1. DNA-PKcs Inhibitors and Radiotherapy

DNA-PKcs inhibitors enhance the efficacy of RT by disrupting DSB repair mechanisms, thereby increasing tumor radiosensitivity. This synergistic approach has demonstrated promising therapeutic potential across multiple preclinical models, including cervical cancer, oral squamous cell carcinoma, non-small cell lung cancer (NSCLC), and gliomas [155,161,162,163]. Beyond impairing DNA repair, DNA-PKcs inhibitors have been shown to modulate the TME under ionizing radiation, suppressing VEGF and HIF-1α expression in glioma cells and impeding microvascular endothelial cell proliferation and metastasis—a finding that opens new avenues for TME-targeted therapies [163].

Peposertib, for instance, exhibits enhanced tumor-targeting specificity when combined with RT, as evidenced in a melanoma brain metastasis model where alterations in DNA-PKcs phosphorylation and subcellular localization were observed [161,164]. Similarly, the dual ATM/DNA-PKcs inhibitor XRD-0394 improves RT response when co-administered with PARP inhibitors or topoisomerase I inhibitors [151]. An ongoing clinical trial (NCT05002140) is further evaluating this combinatorial strategy.

Additionally, the interplay between DNA-PKcs and RT has spurred interest in combining DNA-PKcs inhibitors with radionuclide therapy, with several clinical trials (NCT04071236, NCT04750954) currently investigating this approach [165,166]. These developments highlight the expanding therapeutic potential of DNA-PKcs inhibition in optimizing radiation-based cancer treatments.

Table 2.

Combination Therapies Targeting DNA-PKcs.

Combination Type Mechanism Drugs/Therapies Involved Research Progress Key Findings
Radiotherapy Disrupt DNA repair mechanisms to enhance radiosensitivity Radiotherapy Preclinical animal studies Enhances Radiotherapy efficacy;
expands radiation-based therapy potential;
opens TME-targeted avenues.
Regulate the expression of VEGF and HIF-1α to prevent cancer cell metastasis Radiotherapy Preclinical animal studies
PARP inhibitors or topoisomerase I inhibitors enhance radiosensitivity PARP inhibitors or topoisomerase I inhibitors, Radiotherapy NCT05002140, Phase I
Disrupt DNA repair mechanisms to enhance radiosensitivity Radionuclide therapy NCT04071236, Phase I, Phase II;
NCT04750954, Phase I
Chemotherapy Induce DNA replication stress and impair damage repair pathways Temozolomide Preclinical animal studies Multi-targeted;
synergistic anti-tumor effects;
potential to improve outcomes.
Competitive receptor binding blocks oncogenic signaling Neratinib
Inhibit androgen receptor signaling in prostate cancer Enzalutamide
Weaken DNA repair ability to induce synthetic lethality PARP inhibitors (Niraparib/Olaparib)
Immunotherapy DNA-PKcs overcomes PD-1/PD-L1 resistance by reprogramming TME and regulates PD-L1 expression. Radionuclide therapy, Avelumab NCT04071236, Phase I, Phase II Strong mechanistic basis;
synergizes with checkpoint blockade;
overcome resistance potentially;
potential immunotoxicity.
Radiotherapy, Avelumab NCT04068194, Phase I, Phase II
Novel DNA-PKcs Inhibitor Activating DNA-PK/p53/p21 pathway protects normal cells and enhances cancer cell death. Combined with PARP inhibitors, it induces synthetic lethality in cancer cells. Olaparib NCT05700669, Phase I, Phase II Spares normal tissues, promotes their repair;
low resistance propensity;
synergizes with PARP inhibitors.
Monotherapy NCT03579628, Phase I

4.2.2. DNA-PKcs Inhibitors and Chemotherapy

The therapeutic potential of combining DNA-PKcs inhibitors with conventional chemotherapeutic agents has become an important focus in oncology research. This combination approach exerts synergistic anti-tumor effects through multiple mechanisms of action: (1) Temozolomide induces DNA replication stress and impairs damage repair pathways; (2) Neratinib blocks HER2-mediated oncogenic signaling through competitive receptor binding; (3) Enzalutamide suppresses androgen receptor signaling in prostate cancer; and (4) PARP inhibitors (Niraparib/Olaparib) induce synthetic lethality by compromising DNA repair capacity [111,167,168,169]. These mechanistic insights have driven substantial interest in developing combination regimens incorporating next-generation DNA-PKcs inhibitors like peposertib and AZD7648. Current research efforts are particularly focused on evaluating these agents in combination with both chemotherapy and radiotherapy, with promising preclinical data supporting their potential to overcome treatment resistance and improve therapeutic outcomes [148,152,155,170]. The multi-targeted nature of these combination strategies offers a compelling approach for addressing the complexity of cancer pathogenesis.

4.2.3. DNA-PKcs Inhibitors and Immunotherapy

The multifaceted role of DNA-PKcs in immune regulation, particularly through its involvement in the cGAS-STING signaling pathway, interleukin-mediated responses, PD-L1 expression modulation, and immune cell function regulation, establishes a strong mechanistic foundation for combining DNA-PKcs inhibitors with immunotherapy. The most promising clinical potential appears to lie in combining these inhibitors with PD-1/PD-L1 checkpoint blockade, as evidenced by compelling preclinical data demonstrating synergistic antitumor effects [171]. Recent breakthroughs have elucidated DNA-PKcs’s critical function in macrophage biology, where it orchestrates DNA sensing mechanisms with mTORC2/Rictor/Myc-dependent proliferative signaling, revealing a novel therapeutic avenue for overcoming PD-1/PD-L1 resistance through tumor microenvironment reprogramming [172,173]. Preclinical evaluation of AZD7648 in combination with radiotherapy has demonstrated robust activation of type I interferon responses and significant enhancement of CD8+ T cell-mediated antitumor immunity [154], though careful consideration must be given to potential immunotoxicity profiles [174]. The concurrent modulation of the tumor microenvironment by both DNA-PKcs inhibitors and immunotherapy presents a unique opportunity to potentiate radiotherapy efficacy, prompting the initiation of several clinical trials (NCT04068194, NCT04071236) investigating this triple-combination therapeutic strategy that simultaneously targets DNA damage response pathways and immune checkpoint mechanisms.

4.2.4. Novel DNA-PKcs Inhibitors

Emerging DNA-PKcs inhibitors demonstrate partial selectivity yet still exhibit considerable toxicity to healthy cells, compounded by pharmacokinetic challenges including poor metabolic stability and rapid clearance that have substantially hindered clinical development [164]. Intriguingly, research on AsiDNA has revealed a potentially superior therapeutic strategy—this novel agent uniquely induces G1/S cell cycle arrest in normal epithelial cells and fibroblasts through activation of the DNA-PK/p53/p21 pathway, a protective mechanism absent in p53-deficient tumor cells [157,175,176]. This differential effect not only spares normal tissues during ionizing radiation but actively promotes their DNA repair, making AsiDNA an exceptionally promising candidate for combination radiotherapy. Furthermore, AsiDNA synergizes remarkably with PARP inhibitors, inducing synthetic lethality in tumor cells while demonstrating significantly lower propensity for resistance development compared to conventional therapies [157,160,175,176]. Preclinical studies combining AsiDNA with olaparib have yielded particularly compelling results, showing both reduced toxicity in normal murine cells and enhanced tumor cell death through dual impairment of DNA repair pathways that leads to catastrophic accumulation of unrepaired DNA damage [157,177,178]. These findings have prompted ongoing clinical evaluation (NCT05700669) of this combination in recurrent solid tumors, while a separate trial (NCT03579628) is investigating AsiDNA monotherapy at 600mg in advanced solid tumors, with planned exploration of chemotherapeutic combinations to further potentiate its anticancer efficacy [179]. This next generation of DNA damage response inhibitors represents a paradigm shift in targeted cancer therapy by selectively exploiting tumor-specific vulnerabilities in DNA repair mechanisms.

4.3. Factors Influencing the Efficacy of DNA-PKcs Targeted Therapies

The therapeutic efficacy of DNA-PKcs-targeted interventions is governed by multiple interrelated biological and pharmacological factors. Foremost among these is the substantial variation in PRKDC gene mutation frequencies across cancer types, with colon (9.66%), gastric (9.63%), and endometrial cancers (9.27%) exhibiting markedly higher mutation rates compared to thyroid cancer (0.99%), glioblastoma (1.37%), and hepatocellular carcinoma (1.61%) [180]. Given that PRKDC encodes DNA-PKcs, these mutations can impair V(D)J recombination or kinase activity, with paradoxical effects on therapy response. For instance, kinase-activating mutations (e.g., Thr2609/Ser2056 clusters) may sensitize tumors to DNA-PKcs inhibitors [22,72], whereas truncating mutations could promote resistance. Structural studies further reveal that mutations in the FAT/FATC domains destabilize DNA-PKcs and modulate radiation sensitivity [181], highlighting the need for genomic profiling to guide inhibitor use.

Functional redundancy within the PIKK family further complicates treatment outcomes, as ATM/ATR can partially compensate for DNA-PKcs inhibition through phosphorylation-mediated regulation of downstream effectors [53,182]. Therefore, inhibiting DNA-PKcs, ATM, and ATR together can block the repair pathway and prevent tumor cells from developing drug resistance through compensatory mechanisms [183,184,185]. However, combining PIKK family inhibitors still raises significant safety concerns. The efficacy of ATM/DNA-PKcs co-inhibition is context-dependent, with variable sensitization observed across cell types [185]. Notably, therapeutic response correlates strongly with basal DNA-PKcs activity—low expression may diminish efficacy and promote resistance, independent of inhibitor dosage [182,183].

Emerging evidence suggests that mutations in TP53, ATM, or HRR genes (e.g., BRCA1/2) may predict sensitivity to DNA-PKcs inhibition. For instance, TP53-deficient tumors, reliant on NHEJ due to G1/S checkpoint loss, show heightened vulnerability to DNA-PKcs inhibitors like AZD7648 [154]. Similarly, ATM mutations confer synthetic lethality with DNA-PKcs blockade, as seen in preclinical models of XRD-0394 [151]. HR-deficient cancers (e.g., BRCA-mutated) exhibit synergistic lethality when DNA-PKcs inhibitors are combined with PARP inhibitors [157,177]. Clinical validation of these biomarkers is ongoing in trials such as NCT04071236 (DNA-PKcs + radionuclide therapy) and NCT05002140 (ATM/DNA-PKcs inhibition).

In conclusion, the therapeutic potential of DNA-PKcs inhibition is shaped by complex molecular determinants, including tumor-specific PRKDC mutation profiles, compensatory signaling within the PIKK family, and synergistic interactions with DDR deficiencies. While combination strategies show promise in overcoming resistance, their clinical translation requires careful consideration of biomarker-guided patient selection and toxicity management.

5. Conclusions

Recent research has unveiled DNA-PKcs as a multifunctional regulator in cancer biology, extending well beyond its classical role in NHEJ-mediated DNA repair. The protein’s diverse post-translational modifications enable its involvement in critical processes such as cell cycle regulation, cytotoxic stress responses, immune modulation, and oncogenic progression. Importantly, accumulating evidence strongly correlates heightened DNA-PKcs activity with cancer initiation, progression, and clinical outcomes, solidifying its therapeutic relevance as a molecular target. Although multiple DNA-PKcs inhibitors have entered clinical trials and shown encouraging results-both as monotherapies and in combination regimens—their development has faced substantial challenges.

The current lack of FDA-approved inhibitors stems from suboptimal pharmacokinetics, poor aqueous solubility, and dose-limiting toxicities that have prematurely halted several clinical programs. Moreover, tumor heterogeneity poses a major hurdle for achieving consistent therapeutic responses. The immunosuppressive tumor microenvironment can compromise DNA-PKcs-targeted therapies; for instance, PD-L1 upregulation and impaired immune cell infiltration in triple-negative breast cancer models have been shown to diminish treatment efficacy, though combining DNA-PKcs inhibition with immunotherapy may overcome this limitation. Overcoming these hurdles requires a multipronged approach: deeper mechanistic studies of DNA-PKcs regulation using systems biology methods, longitudinal analyses to identify optimal treatment windows, and rational combination strategies (e.g., with radiotherapy or immunotherapy) to enhance tumor specificity and overcome microenvironmental resistance. Crucially, current inhibitor designs show suboptimal engagement of the catalytic pocket, underscoring the need for structure-based drug optimization. Despite these challenges, the accumulated knowledge provides a strong foundation for developing next-generation inhibitors with improved selectivity and safety profiles, moving DNA-PKcs-targeted therapy closer to clinical application.

Author Contributions

T.Z.: Writing—original draft, formal analysis. C.Y.: investigation, resources. Writing—original draft, conceptualization. C.S.: Software supervision. H.W.: Project administration, funding acquisition. X.L. Writing—review & editing, Supervision, Conceptualization. All authors have read and agreed to the published version of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Funding Statement

This work was jointly supported by the Natural Science Foundation of Gansu Province (No. 25JRRA462), the Isotope Laboratory Self deployment Project of Gansu Province (No. GSTWS2501), the BR Program of Chinese Academy of Sciences (No. E429221YR0) and the Construction and Talent Management of Isotope Manufacturing Innovation Center of Gansu Province (2023RCXM87).

Footnotes

Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

References

  • 1.Yue X., Bai C., Xie D., Ma T., Zhou P.K. DNA-PKcs: A Multi-Faceted Player in DNA Damage Response. Front. Genet. 2020;11:607428. doi: 10.3389/fgene.2020.607428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Wang Y.H., Guo Z., An L., Zhou Y., Xu H., Xiong J., Liu Z.Q., Chen X.P., Zhou H.H., Li X., et al. LINC-PINT impedes DNA repair and enhances radiotherapeutic response by targeting DNA-PKcs in nasopharyngeal cancer. Cell Death Dis. 2021;12:454. doi: 10.1038/s41419-021-03728-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Liu R., Yu X., Chen X., Zhong H., Liang C., Xu X., Xu W., Cheng Y., Wang W., Yu L., et al. Individual factors define the overall effects of dietary genistein exposure on breast cancer patients. Nutr. Res. 2019;67:1–16. doi: 10.1016/j.nutres.2019.03.015. [DOI] [PubMed] [Google Scholar]
  • 4.Rui R., Zhou L., He S. Cancer immunotherapies: Advances and bottlenecks. Front. Immunol. 2023;14:1212476. doi: 10.3389/fimmu.2023.1212476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ruiz-Ceamanos A., Spence C., Navarra J. Individual Differences in Chemosensory Perception Amongst Cancer Patients Undergoing Chemotherapy: A Narrative Review. Nutr. Cancer. 2022;74:1927–1941. doi: 10.1080/01635581.2021.2000625. [DOI] [PubMed] [Google Scholar]
  • 6.Sancar A., Lindsey-Boltz L.A., Unsal-Kacmaz K., Linn S. Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annu. Rev. Biochem. 2004;73:39–85. doi: 10.1146/annurev.biochem.73.011303.073723. [DOI] [PubMed] [Google Scholar]
  • 7.Schirrmacher V. From chemotherapy to biological therapy: A review of novel concepts to reduce the side effects of systemic cancer treatment (Review) Int. J. Oncol. 2019;54:407–419. doi: 10.3892/ijo.2018.4661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lees-Miller S.P., Beattie T.L., Tainer J.A. Noncoding RNA joins Ku and DNA-PKcs for DNA-break resistance in breast cancer. Nat. Struct. Mol. Biol. 2016;23:509–510. doi: 10.1038/nsmb.3240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kidane D., Chae W.J., Czochor J., Eckert K.A., Glazer P.M., Bothwell A.L., Sweasy J.B. Interplay between DNA repair and inflammation, and the link to cancer. Crit. Rev. Biochem. Mol. Biol. 2014;49:116–139. doi: 10.3109/10409238.2013.875514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lee Y.T., Tan Y.J., Oon C.E. Molecular targeted therapy: Treating cancer with specificity. Eur. J. Pharmacol. 2018;834:188–196. doi: 10.1016/j.ejphar.2018.07.034. [DOI] [PubMed] [Google Scholar]
  • 11.Dilalla V., Chaput G., Williams T., Sultanem K. Radiotherapy side effects: Integrating a survivorship clinical lens to better serve patients. Curr. Oncol. 2020;27:107–112. doi: 10.3747/co.27.6233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dagogo-Jack I., Shaw A.T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 2018;15:81–94. doi: 10.1038/nrclinonc.2017.166. [DOI] [PubMed] [Google Scholar]
  • 13.Chang H.H.Y., Pannunzio N.R., Adachi N., Lieber M.R. Non-homologous DNA end joining and alternative pathways to double-strand break repair. Nat. Rev. Mol. Cell Biol. 2017;18:495–506. doi: 10.1038/nrm.2017.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Falck J., Coates J., Jackson S.P. Conserved modes of recruitment of ATM, ATR and DNA-PKcs to sites of DNA damage. Nature. 2005;434:605–611. doi: 10.1038/nature03442. [DOI] [PubMed] [Google Scholar]
  • 15.Douglas P., Sapkota G.P., Morrice N., Yu Y., Goodarzi A.A., Merkle D., Meek K., Alessi D.R., Lees-Miller S.P. Identification of in vitro and in vivo phosphorylation sites in the catalytic subunit of the DNA-dependent protein kinase. Biochem. J. 2002;368:243–251. doi: 10.1042/bj20020973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sibanda B.L., Chirgadze D.Y., Blundell T.L. Crystal structure of DNA-PKcs reveals a large open-ring cradle comprised of HEAT repeats. Nature. 2010;463:118–121. doi: 10.1038/nature08648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gupta S., Meek K. The leucine rich region of DNA-PKcs contributes to its innate DNA affinity. Nucleic Acids Res. 2005;33:6972–6981. doi: 10.1093/nar/gki990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Baretic D., Maia de Oliveira T., Niess M., Wan P., Pollard H., Johnson C.M., Truman C., McCall E., Fisher D., Williams R., et al. Structural insights into the critical DNA damage sensors DNA-PKcs, ATM and ATR. Prog. Biophys. Mol. Biol. 2019;147:4–16. doi: 10.1016/j.pbiomolbio.2019.06.003. [DOI] [PubMed] [Google Scholar]
  • 19.Chen S., Vogt A., Lee L., Naila T., McKeown R., Tomkinson A.E., Lees-Miller S.P., He Y. Cryo-EM visualization of DNA-PKcs structural intermediates in NHEJ. Sci. Adv. 2023;9:eadg2838. doi: 10.1126/sciadv.adg2838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sharif H., Li Y., Dong Y., Dong L., Wang W.L., Mao Y., Wu H. Cryo-EM structure of the DNA-PK holoenzyme. Proc. Natl. Acad. Sci. USA. 2017;114:7367–7372. doi: 10.1073/pnas.1707386114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Jiang W., Crowe J.L., Liu X., Nakajima S., Wang Y., Li C., Lee B.J., Dubois R.L., Liu C., Yu X., et al. Differential phosphorylation of DNA-PKcs regulates the interplay between end-processing and end-ligation during nonhomologous end-joining. Mol. Cell. 2015;58:172–185. doi: 10.1016/j.molcel.2015.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Liu L., Chen X., Li J., Wang H., Buehl C.J., Goff N.J., Meek K., Yang W., Gellert M. Autophosphorylation transforms DNA-PK from protecting to processing DNA ends. Mol. Cell. 2022;82:177–189.e4. doi: 10.1016/j.molcel.2021.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Walker J.R., Corpina R.A., Goldberg J. Structure of the Ku heterodimer bound to DNA and its implications for double-strand break repair. Nature. 2001;412:607–614. doi: 10.1038/35088000. [DOI] [PubMed] [Google Scholar]
  • 24.Spagnolo L., Rivera-Calzada A., Pearl L.H., Llorca O. Three-dimensional structure of the human DNA-PKcs/Ku70/Ku80 complex assembled on DNA and its implications for DNA DSB repair. Mol. Cell. 2006;22:511–519. doi: 10.1016/j.molcel.2006.04.013. [DOI] [PubMed] [Google Scholar]
  • 25.Lieber M.R. The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu. Rev. Biochem. 2010;79:181–211. doi: 10.1146/annurev.biochem.052308.093131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Uematsu N., Weterings E., Yano K., Morotomi-Yano K., Jakob B., Taucher-Scholz G., Mari P.O., van Gent D.C., Chen B.P., Chen D.J. Autophosphorylation of DNA-PKCS regulates its dynamics at DNA double-strand breaks. J. Cell Biol. 2007;177:219–229. doi: 10.1083/jcb.200608077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Dobbs T.A., Tainer J.A., Lees-Miller S.P. A structural model for regulation of NHEJ by DNA-PKcs autophosphorylation. DNA Repair. 2010;9:1307–1314. doi: 10.1016/j.dnarep.2010.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Reddy Y.V., Ding Q., Lees-Miller S.P., Meek K., Ramsden D.A. Non-homologous end joining requires that the DNA-PK complex undergo an autophosphorylation-dependent rearrangement at DNA ends. J. Biol. Chem. 2004;279:39408–39413. doi: 10.1074/jbc.M406432200. [DOI] [PubMed] [Google Scholar]
  • 29.Lu H., Saha J., Beckmann P.J., Hendrickson E.A., Davis A.J. DNA-PKcs promotes chromatin decondensation to facilitate initiation of the DNA damage response. Nucleic Acids Res. 2019;47:9467–9479. doi: 10.1093/nar/gkz694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Rivera-Calzada A., Spagnolo L., Pearl L.H., Llorca O. Structural model of full-length human Ku70-Ku80 heterodimer and its recognition of DNA and DNA-PKcs. EMBO Rep. 2007;8:56–62. doi: 10.1038/sj.embor.7400847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Goodarzi A.A., Yu Y., Riballo E., Douglas P., Walker S.A., Ye R., Harer C., Marchetti C., Morrice N., Jeggo P.A., et al. DNA-PK autophosphorylation facilitates Artemis endonuclease activity. EMBO J. 2006;25:3880–3889. doi: 10.1038/sj.emboj.7601255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Schatz D.G., Swanson P.C. V(D)J recombination: Mechanisms of initiation. Annu. Rev. Genet. 2011;45:167–202. doi: 10.1146/annurev-genet-110410-132552. [DOI] [PubMed] [Google Scholar]
  • 33.Malu S., Malshetty V., Francis D., Cortes P. Role of non-homologous end joining in V(D)J recombination. Immunol. Res. 2012;54:233–246. doi: 10.1007/s12026-012-8329-z. [DOI] [PubMed] [Google Scholar]
  • 34.Ru H., Chambers M.G., Fu T.M., Tong A.B., Liao M., Wu H. Molecular Mechanism of V(D)J Recombination from Synaptic RAG1-RAG2 Complex Structures. Cell. 2015;163:1138–1152. doi: 10.1016/j.cell.2015.10.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Malu S., De Ioannes P., Kozlov M., Greene M., Francis D., Hanna M., Pena J., Escalante C.R., Kurosawa A., Erdjument-Bromage H., et al. Artemis C-terminal region facilitates V(D)J recombination through its interactions with DNA Ligase IV and DNA-PKcs. J. Exp. Med. 2012;209:955–963. doi: 10.1084/jem.20111437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Vu D.D., Bonucci A., Breniere M., Cisneros-Aguirre M., Pelupessy P., Wang Z., Carlier L., Bouvignies G., Cortes P., Aggarwal A.K., et al. Multivalent interactions of the disordered regions of XLF and XRCC4 foster robust cellular NHEJ and drive the formation of ligation-boosting condensates in vitro. Nat. Struct. Mol. Biol. 2024;31:1732–1744. doi: 10.1038/s41594-024-01339-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Xie Y., Liu Y.K., Guo Z.P., Guan H., Liu X.D., Xie D.F., Jiang Y.G., Ma T., Zhou P.K. RBX1 prompts degradation of EXO1 to limit the homologous recombination pathway of DNA double-strand break repair in G1 phase. Cell Death Differ. 2020;27:1383–1397. doi: 10.1038/s41418-019-0424-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Liaw H., Lee D., Myung K. DNA-PK-dependent RPA2 hyperphosphorylation facilitates DNA repair and suppresses sister chromatid exchange. PLoS ONE. 2011;6:e21424. doi: 10.1371/journal.pone.0021424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lu H., Guan J., Wang S.Y., Li G.M., Bohr V.A., Davis A.J. DNA-PKcs-dependent phosphorylation of RECQL4 promotes NHEJ by stabilizing the NHEJ machinery at DNA double-strand breaks. Nucleic Acids Res. 2022;50:5635–5651. doi: 10.1093/nar/gkac375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Justice J.L., Reed T.J., Phelan B., Greco T.M., Hutton J.E., Cristea I.M. DNA-PK and ATM drive phosphorylation signatures that antagonistically regulate cytokine responses to herpesvirus infection or DNA damage. Cell Syst. 2024;15:339–361.e8. doi: 10.1016/j.cels.2024.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hammarsten O., DeFazio L.G., Chu G. Activation of DNA-dependent protein kinase by single-stranded DNA ends. J. Biol. Chem. 2000;275:1541–1550. doi: 10.1074/jbc.275.3.1541. [DOI] [PubMed] [Google Scholar]
  • 42.Zhou Y., Lee J.H., Jiang W., Crowe J.L., Zha S., Paull T.T. Regulation of the DNA Damage Response by DNA-PKcs Inhibitory Phosphorylation of ATM. Mol. Cell. 2017;65:91–104. doi: 10.1016/j.molcel.2016.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lou Z., Chen B.P., Asaithamby A., Minter-Dykhouse K., Chen D.J., Chen J. MDC1 regulates DNA-PK autophosphorylation in response to DNA damage. J. Biol. Chem. 2004;279:46359–46362. doi: 10.1074/jbc.C400375200. [DOI] [PubMed] [Google Scholar]
  • 44.Shang Z.F., Tan W., Liu X.D., Yu L., Li B., Li M., Song M., Wang Y., Xiao B.B., Zhong C.G., et al. DNA-PKcs Negatively Regulates Cyclin B1 Protein Stability through Facilitating Its Ubiquitination Mediated by Cdh1-APC/C Pathway. Int. J. Biol. Sci. 2015;11:1026–1035. doi: 10.7150/ijbs.12443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wang C.Y., Huang E.Y., Huang S.C., Chung B.C. DNA-PK/Chk2 induces centrosome amplification during prolonged replication stress. Oncogene. 2015;34:1263–1269. doi: 10.1038/onc.2014.74. [DOI] [PubMed] [Google Scholar]
  • 46.An J., Yang D.Y., Xu Q.Z., Zhang S.M., Huo Y.Y., Shang Z.F., Wang Y., Wu D.C., Zhou P.K. DNA-dependent protein kinase catalytic subunit modulates the stability of c-Myc oncoprotein. Mol. Cancer. 2008;7:32. doi: 10.1186/1476-4598-7-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Santoni-Rugiu E., Falck J., Mailand N., Bartek J., Lukas J. Involvement of Myc activity in a G(1)/S-promoting mechanism parallel to the pRb/E2F pathway. Mol. Cell Biol. 2000;20:3497–3509. doi: 10.1128/.20.10.3497-3509.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Knudsen K.E., Diehl J.A., Haiman C.A., Knudsen E.S. Cyclin D1: Polymorphism, aberrant splicing and cancer risk. Oncogene. 2006;25:1620–1628. doi: 10.1038/sj.onc.1209371. [DOI] [PubMed] [Google Scholar]
  • 49.Rubin S.M., Sage J., Skotheim J.M. Integrating Old and New Paradigms of G1/S Control. Mol. Cell. 2020;80:183–192. doi: 10.1016/j.molcel.2020.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sakata K., Yamamoto H., Matsumoto Y., Someya M., Hareyama M. cDNA analysis of gene expression associated with DNA-dependent protein kinase activity. Int. J. Oncol. 2007;30:413–420. [PubMed] [Google Scholar]
  • 51.Davis A.J., Chi L., So S., Lee K.J., Mori E., Fattah K., Yang J., Chen D.J. BRCA1 modulates the autophosphorylation status of DNA-PKcs in S phase of the cell cycle. Nucleic Acids Res. 2014;42:11487–11501. doi: 10.1093/nar/gku824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lin Y.F., Shih H.Y., Shang Z., Matsunaga S., Chen B.P. DNA-PKcs is required to maintain stability of Chk1 and Claspin for optimal replication stress response. Nucleic Acids Res. 2014;42:4463–4473. doi: 10.1093/nar/gku116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Mladenov E., Fan X., Dueva R., Soni A., Iliakis G. Radiation-dose-dependent functional synergisms between ATM, ATR and DNA-PKcs in checkpoint control and resection in G(2)-phase. Sci. Rep. 2019;9:8255. doi: 10.1038/s41598-019-44771-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Dibitetto D., Marshall S., Sanchi A., Liptay M., Badar J., Lopes M., Rottenberg S., Smolka M.B. DNA-PKcs promotes fork reversal and chemoresistance. Mol. Cell. 2022;82:3932–3942.e6. doi: 10.1016/j.molcel.2022.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lin Y.F., Shih H.Y., Shang Z.F., Kuo C.T., Guo J., Du C., Lee H., Chen B.P.C. PIDD mediates the association of DNA-PKcs and ATR at stalled replication forks to facilitate the ATR signaling pathway. Nucleic Acids Res. 2018;46:1847–1859. doi: 10.1093/nar/gkx1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Wang J., Sadeghi C.A., Frock R.L. DNA-PKcs suppresses illegitimate chromosome rearrangements. Nucleic Acids Res. 2024;52:5048–5066. doi: 10.1093/nar/gkae140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Xiao M., Zhang S., Liu Z., Mo Y., Wang H., Zhao X., Yang X., Boohaker R.J., Chen Y., Han Y., et al. Dual-functional significance of ATM-mediated phosphorylation of spindle assembly checkpoint component Bub3 in mitosis and the DNA damage response. J. Biol. Chem. 2022;298:101632. doi: 10.1016/j.jbc.2022.101632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lu W., Zhang Y., Liu D., Songyang Z., Wan M. Telomeres-structure, function, and regulation. Exp. Cell Res. 2013;319:133–141. doi: 10.1016/j.yexcr.2012.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Espejel S., Franco S., Sgura A., Gae D., Bailey S.M., Taccioli G.E., Blasco M.A. Functional interaction between DNA-PKcs and telomerase in telomere length maintenance. EMBO J. 2002;21:6275–6287. doi: 10.1093/emboj/cdf593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Sui J., Lin Y.F., Xu K., Lee K.J., Wang D., Chen B.P. DNA-PKcs phosphorylates hnRNP-A1 to facilitate the RPA-to-POT1 switch and telomere capping after replication. Nucleic Acids Res. 2015;43:5971–5983. doi: 10.1093/nar/gkv539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lee H.S., Choe G., Park K.U., Park D.J., Yang H.K., Lee B.L., Kim W.H. Altered expression of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) during gastric carcinogenesis and its clinical implications on gastric cancer. Int. J. Oncol. 2007;31:859–866. doi: 10.3892/ijo.31.4.859. [DOI] [PubMed] [Google Scholar]
  • 62.Woodbine L., Neal J.A., Sasi N.K., Shimada M., Deem K., Coleman H., Dobyns W.B., Ogi T., Meek K., Davies E.G., et al. PRKDC mutations in a SCID patient with profound neurological abnormalities. J. Clin. Investig. 2013;123:2969–2980. doi: 10.1172/JCI67349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Okayasu R., Suetomi K., Yu Y., Silver A., Bedford J.S., Cox R., Ullrich R.L. A deficiency in DNA repair and DNA-PKcs expression in the radiosensitive BALB/c mouse. Cancer Res. 2000;60:4342–4345. [PubMed] [Google Scholar]
  • 64.Yang H., Yao F., Marti T.M., Schmid R.A., Peng R.W. Beyond DNA Repair: DNA-PKcs in Tumor Metastasis, Metabolism and Immunity. Cancers. 2020;12:3389. doi: 10.3390/cancers12113389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ghonim M.A., Pyakurel K., Ju J., Rodriguez P.C., Lammi M.R., Davis C., Abughazleh M.Q., Mansy M.S., Naura A.S., Boulares A.H. DNA-dependent protein kinase inhibition blocks asthma in mice and modulates human endothelial and CD4(+) T-cell function without causing severe combined immunodeficiency. J. Allergy Clin. Immunol. 2015;135:425–440. doi: 10.1016/j.jaci.2014.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wang Y., Fu Z., Li X., Liang Y., Pei S., Hao S., Zhu Q., Yu T., Pei Y., Yuan J., et al. Cytoplasmic DNA sensing by KU complex in aged CD4(+) T cell potentiates T cell activation and aging-related autoimmune inflammation. Immunity. 2021;54:632–647.e639. doi: 10.1016/j.immuni.2021.02.003. [DOI] [PubMed] [Google Scholar]
  • 67.Kim Wiese A., Schluterman Burdine M., Turnage R.H., Tackett A.J., Burdine L.J. DNA-PKcs controls calcineurin mediated IL-2 production in T lymphocytes. PLoS ONE. 2017;12:e0181608. doi: 10.1371/journal.pone.0181608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Waldrip Z.J., Burdine L., Harrison D.K., Azevedo-Pouly A.C., Storey A.J., Moffett O.G., Mackintosh S.G., Burdine M.S. DNA-PKcs kinase activity stabilizes the transcription factor Egr1 in activated immune cells. J. Biol. Chem. 2021;297:101209. doi: 10.1016/j.jbc.2021.101209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Rooney S., Alt F.W., Sekiguchi J., Manis J.P. Artemis-independent functions of DNA-dependent protein kinase in Ig heavy chain class switch recombination and development. Proc. Natl. Acad. Sci. USA. 2005;102:2471–2475. doi: 10.1073/pnas.0409857102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bjorkman A., Du L., Felgentreff K., Rosner C., Pankaj Kamdar R., Kokaraki G., Matsumoto Y., Davies E.G., van der Burg M., Notarangelo L.D., et al. DNA-PKcs Is Involved in Ig Class Switch Recombination in Human B Cells. J. Immunol. 2015;195:5608–5615. doi: 10.4049/jimmunol.1501633. [DOI] [PubMed] [Google Scholar]
  • 71.Crowe J.L., Shao Z., Wang X.S., Wei P.C., Jiang W., Lee B.J., Estes V.M., Alt F.W., Zha S. Kinase-dependent structural role of DNA-PKcs during immunoglobulin class switch recombination. Proc. Natl. Acad. Sci. USA. 2018;115:8615–8620. doi: 10.1073/pnas.1808490115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Crowe J.L., Wang X.S., Shao Z., Lee B.J., Estes V.M., Zha S. DNA-PKcs phosphorylation at the T2609 cluster alters the repair pathway choice during immunoglobulin class switch recombination. Proc. Natl. Acad. Sci. USA. 2020;117:22953–22961. doi: 10.1073/pnas.2007455117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Decout A., Katz J.D., Venkatraman S., Ablasser A. The cGAS-STING pathway as a therapeutic target in inflammatory diseases. Nat. Rev. Immunol. 2021;21:548–569. doi: 10.1038/s41577-021-00524-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chen Q., Sun L., Chen Z.J. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat. Immunol. 2016;17:1142–1149. doi: 10.1038/ni.3558. [DOI] [PubMed] [Google Scholar]
  • 75.Wang F., Zhao M., Chang B., Zhou Y., Wu X., Ma M., Liu S., Cao Y., Zheng M., Dang Y., et al. Cytoplasmic PARP1 links the genome instability to the inhibition of antiviral immunity through PARylating cGAS. Mol. Cell. 2022;82:2032–2049.e2037. doi: 10.1016/j.molcel.2022.03.034. [DOI] [PubMed] [Google Scholar]
  • 76.Liu X., Cen X., Wu R., Chen Z., Xie Y., Wang F., Shan B., Zeng L., Zhou J., Xie B., et al. ARIH1 activates STING-mediated T-cell activation and sensitizes tumors to immune checkpoint blockade. Nat. Commun. 2023;14:4066. doi: 10.1038/s41467-023-39920-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Shao L., Goronzy J.J., Weyand C.M. DNA-dependent protein kinase catalytic subunit mediates T-cell loss in rheumatoid arthritis. EMBO Mol. Med. 2010;2:415–427. doi: 10.1002/emmm.201000096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Zou R., Shi W., Chang X., Zhang M., Tan S., Li R., Zhou H., Li Y., Wang G., Lv W., et al. The DNA-dependent protein kinase catalytic subunit exacerbates endotoxemia-induced myocardial microvascular injury by disrupting the MOTS-c/JNK pathway and inducing profilin-mediated lamellipodia degradation. Theranostics. 2024;14:1561–1582. doi: 10.7150/thno.92650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Fritz G., Kaina B. Late activation of stress kinases (SAPK/JNK) by genotoxins requires the DNA repair proteins DNA-PKcs and CSB. Mol. Biol. Cell. 2006;17:851–861. doi: 10.1091/mbc.e05-07-0606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ghonim M.A., Ju J., Pyakurel K., Ibba S.V., Abouzeid M.M., Rady H.F., Matsuyama S., Del Valle L., Boulares A.H. Unconventional activation of PRKDC by TNF-alpha: Deciphering its crucial role in Th1-mediated inflammation beyond DNA repair as part of the DNA-PK complex. J. Inflamm. 2024;21:14. doi: 10.1186/s12950-024-00386-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ma D., Chen X., Zhang P.Y., Zhang H., Wei L.J., Hu S., Tang J.Z., Zhou M.T., Xie C., Ou R., et al. Upregulation of the ALDOA/DNA-PK/p53 pathway by dietary restriction suppresses tumor growth. Oncogene. 2018;37:1041–1048. doi: 10.1038/onc.2017.398. [DOI] [PubMed] [Google Scholar]
  • 82.Gladdy R.A., Nutter L.M., Kunath T., Danska J.S., Guidos C.J. p53-Independent apoptosis disrupts early organogenesis in embryos lacking both ataxia-telangiectasia mutated and Prkdc. Mol. Cancer Res. 2006;4:311–318. doi: 10.1158/1541-7786.MCR-05-0258. [DOI] [PubMed] [Google Scholar]
  • 83.Fedier A., Moawad A., Haller U., Fink D. p53-deficient cells display increased sensitivity to anthracyclines after loss of the catalytic subunit of the DNA-dependent protein kinase. Int. J. Oncol. 2003;23:1431–1437. doi: 10.3892/ijo.23.5.1431. [DOI] [PubMed] [Google Scholar]
  • 84.Butte M.J., Keir M.E., Phamduy T.B., Sharpe A.H., Freeman G.J. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity. 2007;27:111–122. doi: 10.1016/j.immuni.2007.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Tu X., Qin B., Zhang Y., Zhang C., Kahila M., Nowsheen S., Yin P., Yuan J., Pei H., Li H., et al. PD-L1 (B7-H1) Competes with the RNA Exosome to Regulate the DNA Damage Response and Can Be Targeted to Sensitize to Radiation or Chemotherapy. Mol. Cell. 2019;74:1215–1226.e1214. doi: 10.1016/j.molcel.2019.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zhang Y., Shen G., Meng T., Lv Z., Li X., Li J., Li K. Eicosapentaenoic acid enhances the sensitivity of osteosarcoma to cisplatin by inducing ferroptosis through the DNA-PKcs/AKT/NRF2 pathway and reducing PD-L1 expression to attenuate immune evasion. Int. Immunopharmacol. 2023;125:111181. doi: 10.1016/j.intimp.2023.111181. [DOI] [PubMed] [Google Scholar]
  • 87.Ho S.R., Lee Y.C., Ittmann M.M., Lin F.T., Chan K.S., Lin W.C. RNF144A deficiency promotes PD-L1 protein stabilization and carcinogen-induced bladder tumorigenesis. Cancer Lett. 2021;520:344–360. doi: 10.1016/j.canlet.2021.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Miao Z., Li J., Wang Y., Shi M., Gu X., Zhang X., Wei F., Tang X., Zheng L., Xing Y. Hsa_circ_0136666 stimulates gastric cancer progression and tumor immune escape by regulating the miR-375/PRKDC Axis and PD-L1 phosphorylation. Mol. Cancer. 2023;22:205. doi: 10.1186/s12943-023-01883-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ho S.R., Mahanic C.S., Lee Y.J., Lin W.C. RNF144A, an E3 ubiquitin ligase for DNA-PKcs, promotes apoptosis during DNA damage. Proc. Natl. Acad. Sci. USA. 2014;111:E2646–E2655. doi: 10.1073/pnas.1323107111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sheppard H.M., Liu X. Transcription by RNA polymerase II in DNA-PK deficient scid mouse cells. Biochim. Biophys. Acta. 2000;1493:41–47. doi: 10.1016/S0167-4781(00)00155-X. [DOI] [PubMed] [Google Scholar]
  • 91.Koh M.Y., Lemos R., Jr., Liu X., Powis G. The hypoxia-associated factor switches cells from HIF-1alpha- to HIF-2alpha-dependent signaling promoting stem cell characteristics, aggressive tumor growth and invasion. Cancer Res. 2011;71:4015–4027. doi: 10.1158/0008-5472.CAN-10-4142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Vanichapol T., Leelawat K., Hongeng S. Hypoxia enhances cholangiocarcinoma invasion through activation of hepatocyte growth factor receptor and the extracellular signal-regulated kinase signaling pathway. Mol. Med. Rep. 2015;12:3265–3272. doi: 10.3892/mmr.2015.3865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Cowman S.J., Koh M.Y. Revisiting the HIF switch in the tumor and its immune microenvironment. Trends Cancer. 2022;8:28–42. doi: 10.1016/j.trecan.2021.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lee S.H., Golinska M., Griffiths J.R. HIF-1-Independent Mechanisms Regulating Metabolic Adaptation in Hypoxic Cancer Cells. Cells. 2021;10:2371. doi: 10.3390/cells10092371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Yang Y., Lu H., Chen C., Lyu Y., Cole R.N., Semenza G.L. HIF-1 Interacts with TRIM28 and DNA-PK to release paused RNA polymerase II and activate target gene transcription in response to hypoxia. Nat. Commun. 2022;13:316. doi: 10.1038/s41467-021-27944-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Li P., Gai X., Li Q., Yang Q., Yu X. DNA-PK participates in pre-rRNA biogenesis independent of DNA double-strand break repair. Nucleic Acids Res. 2024;52:6360–6375. doi: 10.1093/nar/gkae316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Zhang S., Schlott B., Gorlach M., Grosse F. DNA-dependent protein kinase (DNA-PK) phosphorylates nuclear DNA helicase II/RNA helicase A and hnRNP proteins in an RNA-dependent manner. Nucleic Acids Res. 2004;32:1–10. doi: 10.1093/nar/gkg933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wong R.H., Chang I., Hudak C.S., Hyun S., Kwan H.Y., Sul H.S. A role of DNA-PK for the metabolic gene regulation in response to insulin. Cell. 2009;136:1056–1072. doi: 10.1016/j.cell.2008.12.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Nock A., Ascano J.M., Jones T., Barrero M.J., Sugiyama N., Tomita M., Ishihama Y., Malik S. Identification of DNA-dependent protein kinase as a cofactor for the forkhead transcription factor FoxA2. J. Biol. Chem. 2009;284:19915–19926. doi: 10.1074/jbc.M109.016295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hasegawa Y., Asada S. DNA-dependent protein kinase catalytic subunit binds to the transactivation domain 1 of NF-kappaB p65. Biochem. Biophys. Rep. 2023;35:101538. doi: 10.1016/j.bbrep.2023.101538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Wei S.J., Yang I.H., Mohiuddin I.S., Kshirsagar G.J., Nguyen T.H., Trasti S., Maurer B.J., Kang M.H. DNA-PKcs as an upstream mediator of OCT4-induced MYC activation in small cell lung cancer. Biochim. Biophys. Acta Gene Regul. Mech. 2023;1866:194939. doi: 10.1016/j.bbagrm.2023.194939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hosoi Y., Watanabe T., Nakagawa K., Matsumoto Y., Enomoto A., Morita A., Nagawa H., Suzuki N. Up-regulation of DNA-dependent protein kinase activity and Sp1 in colorectal cancer. Int. J. Oncol. 2004;25:461–468. doi: 10.3892/ijo.25.2.461. [DOI] [PubMed] [Google Scholar]
  • 103.An J., Xu Q.Z., Sui J.L., Bai B., Zhou P.K. Silencing of DNA-PKcs alters the transcriptional profile of certain signal transduction genes related to proliferation and differentiation in HeLa cells. Int. J. Mol. Med. 2005;16:455–462. doi: 10.3892/ijmm.16.3.455. [DOI] [PubMed] [Google Scholar]
  • 104.Wang S., Zhu H., Li R., Mui D., Toan S., Chang X., Zhou H. DNA-PKcs interacts with and phosphorylates Fis1 to induce mitochondrial fragmentation in tubular cells during acute kidney injury. Sci. Signal. 2022;15:eabh1121. doi: 10.1126/scisignal.abh1121. [DOI] [PubMed] [Google Scholar]
  • 105.Mollinari C., Cardinale A., Lupacchini L., Martire A., Chiodi V., Martinelli A., Rinaldi A.M., Fini M., Pazzaglia S., Domenici M.R., et al. The DNA repair protein DNA-PKcs modulates synaptic plasticity via PSD-95 phosphorylation and stability. EMBO Rep. 2024;25:3707–3737. doi: 10.1038/s44319-024-00198-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Du Y., Zhu P., Li Y., Yu J., Xia T., Chang X., Zhu H., Li R., He Q. DNA-PKcs Phosphorylates Cofilin2 to Induce Endothelial Dysfunction and Microcirculatory Disorder in Endotoxemic Cardiomyopathy. Research. 2024;7:0331. doi: 10.34133/research.0331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Adamson B., Brittain N., Walker L., Duncan R., Luzzi S., Rescigno P., Smith G., McGill S., Burchmore R.J., Willmore E., et al. The catalytic subunit of DNA-PK regulates transcription and splicing of AR in advanced prostate cancer. J. Clin. Investig. 2023;133:e169200. doi: 10.1172/JCI169200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Liu L., Deng Y., Zheng Z., Deng Z., Zhang J., Li J., Liang M., Zhou X., Tan W., Yang H., et al. Hsp90 Inhibitor STA9090 Sensitizes Hepatocellular Carcinoma to Hyperthermia-Induced DNA Damage by Suppressing DNA-PKcs Protein Stability and mRNA Transcription. Mol. Cancer Ther. 2021;20:1880–1892. doi: 10.1158/1535-7163.MCT-21-0215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hashimoto T., Urushihara Y., Murata Y., Fujishima Y., Hosoi Y. AMPK increases expression of ATM through transcriptional factor Sp1 and induces radioresistance under severe hypoxia in glioblastoma cell lines. Biochem. Biophys. Res. Commun. 2022;590:82–88. doi: 10.1016/j.bbrc.2021.12.076. [DOI] [PubMed] [Google Scholar]
  • 110.Hu H., Gu Y., Qian Y., Hu B., Zhu C., Wang G., Li J. DNA-PKcs is important for Akt activation and gemcitabine resistance in PANC-1 pancreatic cancer cells. Biochem. Biophys. Res. Commun. 2014;452:106–111. doi: 10.1016/j.bbrc.2014.08.059. [DOI] [PubMed] [Google Scholar]
  • 111.Tomar M.S., Kumar A., Srivastava C., Shrivastava A. Elucidating the mechanisms of Temozolomide resistance in gliomas and the strategies to overcome the resistance. Biochim. Biophys. Acta Rev. Cancer. 2021;1876:188616. doi: 10.1016/j.bbcan.2021.188616. [DOI] [PubMed] [Google Scholar]
  • 112.Zhou Y., Liu F., Xu Q., Yang B., Li X., Jiang S., Hu L., Zhang X., Zhu L., Li Q., et al. Inhibiting Importin 4-mediated nuclear import of CEBPD enhances chemosensitivity by repression of PRKDC-driven DNA damage repair in cervical cancer. Oncogene. 2020;39:5633–5648. doi: 10.1038/s41388-020-1384-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Yan D., Ng W.L., Zhang X., Wang P., Zhang Z., Mo Y.Y., Mao H., Hao C., Olson J.J., Curran W.J., et al. Targeting DNA-PKcs and ATM with miR-101 sensitizes tumors to radiation. PLoS ONE. 2010;5:e11397. doi: 10.1371/journal.pone.0011397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Guo Z., Wang Y.H., Xu H., Yuan C.S., Zhou H.H., Huang W.H., Wang H., Zhang W. LncRNA linc00312 suppresses radiotherapy resistance by targeting DNA-PKcs and impairing DNA damage repair in nasopharyngeal carcinoma. Cell Death Dis. 2021;12:69. doi: 10.1038/s41419-020-03302-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Bergstrand S., O’Brien E.M., Coucoravas C., Hrossova D., Peirasmaki D., Schmidli S., Dhanjal S., Pederiva C., Siggens L., Mortusewicz O., et al. Small Cajal body-associated RNA 2 (scaRNA2) regulates DNA repair pathway choice by inhibiting DNA-PK. Nat. Commun. 2022;13:1015. doi: 10.1038/s41467-022-28646-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Zhang Y., He Q., Hu Z., Feng Y., Fan L., Tang Z., Yuan J., Shan W., Li C., Hu X., et al. Long noncoding RNA LINP1 regulates repair of DNA double-strand breaks in triple-negative breast cancer. Nat. Struct. Mol. Biol. 2016;23:522–530. doi: 10.1038/nsmb.3211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Chen H., Pei L., Xie P., Guo G. Circ-PRKDC Contributes to 5-Fluorouracil Resistance of Colorectal Cancer Cells by Regulating miR-375/FOXM1 Axis and Wnt/beta-Catenin Pathway. OncoTargets Ther. 2020;13:5939–5953. doi: 10.2147/OTT.S253468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Wang G., Li Y., Zhu H., Huo G., Bai J., Gao Z. Circ-PRKDC Facilitates the Progression of Colorectal Cancer Through miR-198/DDR1 Regulatory Axis. Cancer Manag. Res. 2020;12:12853–12865. doi: 10.2147/CMAR.S273484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Iliakis G.E. New Players in the Regulation of DNA-PK Activity: Survivin Joins the Crowd. Cancer Res. 2021;81:2270–2271. doi: 10.1158/0008-5472.CAN-21-0273. [DOI] [PubMed] [Google Scholar]
  • 120.Gullulu O., Hehlgans S., Mayer B.E., Gossner I., Petraki C., Hoffmann M., Dombrowsky M.J., Kunzmann P., Hamacher K., Strebhardt K., et al. A Spatial and Functional Interaction of a Heterotetramer Survivin-DNA-PKcs Complex in DNA Damage Response. Cancer Res. 2021;81:2304–2317. doi: 10.1158/0008-5472.CAN-20-2931. [DOI] [PubMed] [Google Scholar]
  • 121.Lee K.J., Lin Y.F., Chou H.Y., Yajima H., Fattah K.R., Lee S.C., Chen B.P. Involvement of DNA-dependent protein kinase in normal cell cycle progression through mitosis. J. Biol. Chem. 2011;286:12796–12802. doi: 10.1074/jbc.M110.212969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Chen B.P., Uematsu N., Kobayashi J., Lerenthal Y., Krempler A., Yajima H., Lobrich M., Shiloh Y., Chen D.J. Ataxia telangiectasia mutated (ATM) is essential for DNA-PKcs phosphorylations at the Thr-2609 cluster upon DNA double strand break. J. Biol. Chem. 2007;282:6582–6587. doi: 10.1074/jbc.M611605200. [DOI] [PubMed] [Google Scholar]
  • 123.Dylgjeri E., Kothari V., Shafi A.A., Semenova G., Gallagher P.T., Guan Y.F., Pang A., Goodwin J.F., Irani S., McCann J.J., et al. A Novel Role for DNA-PK in Metabolism by Regulating Glycolysis in Castration-Resistant Prostate Cancer. Clin. Cancer Res. 2022;28:1446–1459. doi: 10.1158/1078-0432.CCR-21-1846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Li C., Liu X., Liu Y., Liu X., Wang R., Liao J., Wu S., Fan J., Peng Z., Li B., et al. Keratin 80 promotes migration and invasion of colorectal carcinoma by interacting with PRKDC via activating the AKT pathway. Cell Death Dis. 2018;9:1009. doi: 10.1038/s41419-018-1030-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Zhang H., Ma R.R., Zhang G., Dong Y., Duan M., Sun Y., Tian Y., Gao J.W., Chen X., Liu H.T., et al. Long noncoding RNA lnc-LEMGC combines with DNA-PKcs to suppress gastric cancer metastasis. Cancer Lett. 2022;524:82–90. doi: 10.1016/j.canlet.2021.09.042. [DOI] [PubMed] [Google Scholar]
  • 126.Pan Y., Zhu Q., Hong T., Cheng J., Tang X. Targeting PRKDC activates the efficacy of antitumor immunity while sensitizing to chemotherapy and targeted therapy in liver hepatocellular carcinoma. Aging. 2024;16:9047–9071. doi: 10.18632/aging.205855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Zhang J., Jiang H., Xu D., Wu W.J., Chen H.D., He L. DNA-PKcs Mediates an Epithelial-Mesenchymal Transition Process Promoting Cutaneous Squamous Cell Carcinoma Invasion and Metastasis by Targeting The TGF-beta1/Smad Signaling Pathway. OncoTargets Ther. 2019;12:9395–9405. doi: 10.2147/OTT.S205017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Xu C., Chen G., Yu B., Sun B., Zhang Y., Zhang M., Yang Y., Xiao Y., Cheng S.Y., Li Y., et al. TRIM24 Cooperates with Ras Mutation to Drive Glioma Progression through snoRNA Recruitment of PHAX and DNA-PKcs. Adv. Sci. 2024;11:e2400023. doi: 10.1002/advs.202400023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Wang L., Wu L., Du Y., Wang X., Yang B., Guo S., Zhou Y., Xu Y., Yang S., Zhang Y., et al. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) drives angiotensin II-induced vascular remodeling through regulating mitochondrial fragmentation. Redox Biol. 2023;67:102893. doi: 10.1016/j.redox.2023.102893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Rahal O.N., Fatfat M., Hankache C., Osman B., Khalife H., Machaca K., Muhtasib H.G. Chk1 and DNA-PK mediate TPEN-induced DNA damage in a ROS dependent manner in human colon cancer cells. Cancer Biol. Ther. 2016;17:1139–1148. doi: 10.1080/15384047.2016.1235658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Fan Y., He S. The Characteristics of Tumor Microenvironment in Triple Negative Breast Cancer. Cancer Manag. Res. 2022;14:1–17. doi: 10.2147/CMAR.S316700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Ma C., Qin Y., Wang Y., Zhu C., Gao X., Zhang P., Gu Y., Zhang S., Lin J., Wang J., et al. Targeting DNA-PKcs promotes anti-tumoral immunity via triggering cytosolic DNA sensing and inducing an inflamed tumor immuno-microenvironment in metastatic triple-negative breast cancer. Genes. Dis. 2023;10:1809–1811. doi: 10.1016/j.gendis.2023.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Liu X., Wang Q., Liu B., Zheng X., Li P., Zhao T., Jin X., Ye F., Zhang P., Chen W., et al. Genistein inhibits radiation-induced invasion and migration of glioblastoma cells by blocking the DNA-PKcs/Akt2/Rac1 signaling pathway. Radiother. Oncol. 2021;155:93–104. doi: 10.1016/j.radonc.2020.10.026. [DOI] [PubMed] [Google Scholar]
  • 134.Han D., Liu W., Li G., Liu L. Circ_PRKDC knockdown promotes skin wound healing by enhancing keratinocyte migration via miR-31/FBN1 axis. J. Mol. Histol. 2021;52:681–691. doi: 10.1007/s10735-021-09996-8. [DOI] [PubMed] [Google Scholar]
  • 135.Lee H.S., Yang H.K., Kim W.H., Choe G. Loss of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) expression in gastric cancers. Cancer Res. Treat. 2005;37:98–102. doi: 10.4143/crt.2005.37.2.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Tan J., Sun X., Zhao H., Guan H., Gao S., Zhou P.K. Double-strand DNA break repair: Molecular mechanisms and therapeutic targets. MedComm. 2023;4:e388. doi: 10.1002/mco2.388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Timme C.R., Rath B.H., O’Neill J.W., Camphausen K., Tofilon P.J. The DNA-PK Inhibitor VX-984 Enhances the Radiosensitivity of Glioblastoma Cells Grown In Vitro and as Orthotopic Xenografts. Mol. Cancer Ther. 2018;17:1207–1216. doi: 10.1158/1535-7163.MCT-17-1267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Talele S., Zhang W., Oh J.H., Burgenske D.M., Mladek A.C., Dragojevic S., Sarkaria J.N., Elmquist W.F. Central Nervous System Delivery of the Catalytic Subunit of DNA-Dependent Protein Kinase Inhibitor Peposertib as Radiosensitizer for Brain Metastases. J. Pharmacol. Exp. Ther. 2022;381:217–228. doi: 10.1124/jpet.121.001069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Ohuchi K., Saga R., Hasegawa K., Tsuruga E., Hosokawa Y., Fukumoto M., Okumura K. DNA-PKcs phosphorylation specific inhibitor, NU7441, enhances the radiosensitivity of clinically relevant radioresistant oral squamous cell carcinoma cells. Biomed. Rep. 2023;18:28. doi: 10.3892/br.2023.1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Liang S., Thomas S.E., Chaplin A.K., Hardwick S.W., Chirgadze D.Y., Blundell T.L. Structural insights into inhibitor regulation of the DNA repair protein DNA-PKcs. Nature. 2022;601:643–648. doi: 10.1038/s41586-021-04274-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Nutley B.P., Smith N.F., Hayes A., Kelland L.R., Brunton L., Golding B.T., Smith G.C., Martin N.M., Workman P., Raynaud F.I. Preclinical pharmacokinetics and metabolism of a novel prototype DNA-PK inhibitor NU7026. Br. J. Cancer. 2005;93:1011–1018. doi: 10.1038/sj.bjc.6602823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Zhao Y., Thomas H.D., Batey M.A., Cowell I.G., Richardson C.J., Griffin R.J., Calvert A.H., Newell D.R., Smith G.C., Curtin N.J. Preclinical evaluation of a potent novel DNA-dependent protein kinase inhibitor NU7441. Cancer Res. 2006;66:5354–5362. doi: 10.1158/0008-5472.CAN-05-4275. [DOI] [PubMed] [Google Scholar]
  • 143.Ciszewski W.M., Tavecchio M., Dastych J., Curtin N.J. DNA-PK inhibition by NU7441 sensitizes breast cancer cells to ionizing radiation and doxorubicin. Breast Cancer Res. Treat. 2014;143:47–55. doi: 10.1007/s10549-013-2785-6. [DOI] [PubMed] [Google Scholar]
  • 144.Willmore E., de Caux S., Sunter N.J., Tilby M.J., Jackson G.H., Austin C.A., Durkacz B.W. A novel DNA-dependent protein kinase inhibitor, NU7026, potentiates the cytotoxicity of topoisomerase II poisons used in the treatment of leukemia. Blood. 2004;103:4659–4665. doi: 10.1182/blood-2003-07-2527. [DOI] [PubMed] [Google Scholar]
  • 145.van Bussel M.T.J., Awada A., de Jonge M.J.A., Mau-Sorensen M., Nielsen D., Schoffski P., Verheul H.M.W., Sarholz B., Berghoff K., El Bawab S., et al. A first-in-man phase 1 study of the DNA-dependent protein kinase inhibitor peposertib (formerly M3814) in patients with advanced solid tumours. Br. J. Cancer. 2021;124:728–735. doi: 10.1038/s41416-020-01151-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Chen F., Zhao H., Li C., Li P., Zhang Q. An mTOR and DNA-PK dual inhibitor CC-115 hinders non-small cell lung cancer cell growth. Cell Death Discov. 2022;8:293. doi: 10.1038/s41420-022-01082-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Tsuji T., Sapinoso L.M., Tran T., Gaffney B., Wong L., Sankar S., Raymon H.K., Mortensen D.S., Xu S. CC-115, a dual inhibitor of mTOR kinase and DNA-PK, blocks DNA damage repair pathways and selectively inhibits ATM-deficient cell growth in vitro. Oncotarget. 2017;8:74688–74702. doi: 10.18632/oncotarget.20342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Samuels M., Falkenius J., Bar-Ad V., Dunst J., van Triest B., Yachnin J., Rodriguez-Gutierrez A., Kuipers M., You X., Sarholz B., et al. A Phase 1 Study of the DNA-PK Inhibitor Peposertib in Combination with Radiation Therapy With or Without Cisplatin in Patients with Advanced Head and Neck Tumors. Int. J. Radiat. Oncol. Biol. Phys. 2024;118:743–756. doi: 10.1016/j.ijrobp.2023.09.024. [DOI] [PubMed] [Google Scholar]
  • 149.Munster P., Mita M., Mahipal A., Nemunaitis J., Massard C., Mikkelsen T., Cruz C., Paz-Ares L., Hidalgo M., Rathkopf D., et al. First-In-Human Phase I Study of A Dual mTOR Kinase And DNA-PK Inhibitor (CC-115) In Advanced Malignancy. Cancer Manag. Res. 2019;11:10463–10476. doi: 10.2147/CMAR.S208720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Zhao J.L., Antonarakis E.S., Cheng H.H., George D.J., Aggarwal R., Riedel E., Sumiyoshi T., Schonhoft J.D., Anderson A., Mao N., et al. Phase 1b study of enzalutamide plus CC-115, a dual mTORC1/2 and DNA-PK inhibitor, in men with metastatic castration-resistant prostate cancer (mCRPC) Br. J. Cancer. 2024;130:53–62. doi: 10.1038/s41416-023-02487-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Gilmer T.M., Lai C.H., Guo K., Deland K., Ashcraft K.A., Stewart A.E., Wang Y., Fu J., Wood K.C., Kirsch D.G., et al. A Novel Dual ATM/DNA-PK Inhibitor, XRD-0394, Potently Radiosensitizes and Potentiates PARP and Topoisomerase I Inhibitors. Mol. Cancer Ther. 2024;23:751–765. doi: 10.1158/1535-7163.MCT-23-0890. [DOI] [PubMed] [Google Scholar]
  • 152.Anastasia A., Dellavedova G., Ramos-Montoya A., James N., Chiorino G., Russo M., Baakza H., Wilson J., Ghilardi C., Cadogan E.B., et al. The DNA-PK Inhibitor AZD7648 Sensitizes Patient-Derived Ovarian Cancer Xenografts to Pegylated Liposomal Doxorubicin and Olaparib Preventing Abdominal Metastases. Mol. Cancer Ther. 2022;21:555–567. doi: 10.1158/1535-7163.MCT-21-0420. [DOI] [PubMed] [Google Scholar]
  • 153.Ha J., Park M., Lee Y., Choi S.H., Kim B.S., Ha H., Jeong Y.K. AZD7648, a DNA-PKcs inhibitor, overcomes radioresistance in Hep3B xenografts and cells under tumor hypoxia. Am. J. Cancer Res. 2023;13:4918–4930. [PMC free article] [PubMed] [Google Scholar]
  • 154.Nakamura K., Karmokar A., Farrington P.M., James N.H., Ramos-Montoya A., Bickerton S.J., Hughes G.D., Illidge T.M., Cadogan E.B., Davies B.R., et al. Inhibition of DNA-PK with AZD7648 Sensitizes Tumor Cells to Radiotherapy and Induces Type I IFN-Dependent Durable Tumor Control. Clin. Cancer Res. 2021;27:4353–4366. doi: 10.1158/1078-0432.CCR-20-3701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Fok J.H.L., Ramos-Montoya A., Vazquez-Chantada M., Wijnhoven P.W.G., Follia V., James N., Farrington P.M., Karmokar A., Willis S.E., Cairns J., et al. AZD7648 is a potent and selective DNA-PK inhibitor that enhances radiation, chemotherapy and olaparib activity. Nat. Commun. 2019;10:5065. doi: 10.1038/s41467-019-12836-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Khan A.J., Misenko S.M., Thandoni A., Schiff D., Jhawar S.R., Bunting S.F., Haffty B.G. VX-984 is a selective inhibitor of non-homologous end joining, with possible preferential activity in transformed cells. Oncotarget. 2018;9:25833–25841. doi: 10.18632/oncotarget.25383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Jdey W., Thierry S., Russo C., Devun F., Al Abo M., Noguiez-Hellin P., Sun J.S., Barillot E., Zinovyev A., Kuperstein I., et al. Drug-Driven Synthetic Lethality: Bypassing Tumor Cell Genetics with a Combination of AsiDNA and PARP Inhibitors. Clin. Cancer Res. 2017;23:1001–1011. doi: 10.1158/1078-0432.CCR-16-1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Quanz M., Berthault N., Roulin C., Roy M., Herbette A., Agrario C., Alberti C., Josserand V., Coll J.L., Sastre-Garau X., et al. Small-molecule drugs mimicking DNA damage: A new strategy for sensitizing tumors to radiotherapy. Clin. Cancer Res. 2009;15:1308–1316. doi: 10.1158/1078-0432.CCR-08-2108. [DOI] [PubMed] [Google Scholar]
  • 159.Croset A., Cordelieres F.P., Berthault N., Buhler C., Sun J.S., Quanz M., Dutreix M. Inhibition of DNA damage repair by artificial activation of PARP with siDNA. Nucleic Acids Res. 2013;41:7344–7355. doi: 10.1093/nar/gkt522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Jdey W., Kozlak M., Alekseev S., Thierry S., Lascaux P., Girard P.M., Bono F., Dutreix M. AsiDNA Treatment Induces Cumulative Antitumor Efficacy with a Low Probability of Acquired Resistance. Neoplasia. 2019;21:863–871. doi: 10.1016/j.neo.2019.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Gordhandas S.B., Manning-Geist B., Henson C., Iyer G., Gardner G.J., Sonoda Y., Moore K.N., Aghajanian C., Chui M.H., Grisham R.N. Pre-clinical activity of the oral DNA-PK inhibitor, peposertib (M3814), combined with radiation in xenograft models of cervical cancer. Sci. Rep. 2022;12:974. doi: 10.1038/s41598-021-04618-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Baker J.H.E., Kyle A.H., Liu N.A., Wang T., Liu X., Teymori S., Banath J.P., Minchinton A.I. Radiation and Chemo-Sensitizing Effects of DNA-PK Inhibitors Are Proportional in Tumors and Normal Tissues. Mol. Cancer Ther. 2024;23:1230–1240. doi: 10.1158/1535-7163.MCT-23-0681. [DOI] [PubMed] [Google Scholar]
  • 163.Liu Y., Zhang L., Liu Y., Sun C., Zhang H., Miao G., Di C.X., Zhou X., Zhou R., Wang Z. DNA-PKcs deficiency inhibits glioblastoma cell-derived angiogenesis after ionizing radiation. J. Cell Physiol. 2015;230:1094–1103. doi: 10.1002/jcp.24841. [DOI] [PubMed] [Google Scholar]
  • 164.Ji J., Dragojevic S., Callaghan C.M., Smith E.J., Talele S., Zhang W., Connors M.A., Mladek A.C., Hu Z., Bakken K.K., et al. Differential Distribution of the DNA-PKcs Inhibitor Peposertib Selectively Radiosensitizes Patient-derived Melanoma Brain Metastasis Xenografts. Mol. Cancer Ther. 2024;23:662–671. doi: 10.1158/1535-7163.MCT-23-0552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Waldeck K., Van Zuylekom J., Cullinane C., Gulati T., Simpson K.J., Tothill R.W., Blyth B., Hicks R.J. A genome-wide CRISPR/Cas9 screen identifies DNA-PK as a sensitiser to (177)Lutetium-DOTA-octreotate radionuclide therapy. Theranostics. 2023;13:4745–4761. doi: 10.7150/thno.84628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Reuvers T.G.A., Verkaik N.S., Stuurman D., de Ridder C., Groningen M.C.C., de Blois E., Nonnekens J. DNA-PKcs inhibitors sensitize neuroendocrine tumor cells to peptide receptor radionuclide therapy in vitro and in vivo. Theranostics. 2023;13:3117–3130. doi: 10.7150/thno.82963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Hussain M., Fizazi K., Saad F., Rathenborg P., Shore N., Ferreira U., Ivashchenko P., Demirhan E., Modelska K., Phung D., et al. Enzalutamide in Men with Nonmetastatic, Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2018;378:2465–2474. doi: 10.1056/NEJMoa1800536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Guo L., Shao W., Zhou C., Yang H., Yang L., Cai Q., Wang J., Shi Y., Huang L., Zhang J. Neratinib for HER2-positive breast cancer with an overlooked option. Mol. Med. 2023;29:134. doi: 10.1186/s10020-023-00736-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.D’Andrea A.D. Mechanisms of PARP inhibitor sensitivity and resistance. DNA Repair. 2018;71:172–176. doi: 10.1016/j.dnarep.2018.08.021. [DOI] [PubMed] [Google Scholar]
  • 170.Perez B., Aljumaily R., Marron T.U., Shafique M.R., Burris H., Iams W.T., Chmura S.J., Luke J.J., Edenfield W., Sohal D., et al. Phase I study of peposertib and avelumab with or without palliative radiotherapy in patients with advanced solid tumors. ESMO Open. 2024;9:102217. doi: 10.1016/j.esmoop.2023.102217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Tang Q., Chen Y., Li X., Long S., Shi Y., Yu Y., Wu W., Han L., Wang S. The role of PD-1/PD-L1 and application of immune-checkpoint inhibitors in human cancers. Front. Immunol. 2022;13:964442. doi: 10.3389/fimmu.2022.964442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Gerlach B.D., Ampomah P.B., Yurdagul A., Jr., Liu C., Lauring M.C., Wang X., Kasikara C., Kong N., Shi J., Tao W., et al. Efferocytosis induces macrophage proliferation to help resolve tissue injury. Cell Metab. 2021;33:2445–2463.e8. doi: 10.1016/j.cmet.2021.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Zhang H., Liu L., Liu J., Dang P., Hu S., Yuan W., Sun Z., Liu Y., Wang C. Roles of tumor-associated macrophages in anti-PD-1/PD-L1 immunotherapy for solid cancers. Mol. Cancer. 2023;22:58. doi: 10.1186/s12943-023-01725-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Azevedo-Pouly A.C., Appell L.E., Burdine L., Rogers L.J., Morehead L.C., Fil D., Barker M., Rainwater R.R., Waldrip Z.J., Koss B., et al. Chemical inhibition of DNA-PKcs impairs the activation and cytotoxicity of CD4(+) helper and CD8(+) effector T cells. Immunol. Cell Biol. 2023;101:663–671. doi: 10.1111/imcb.12651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Sesink A., Becerra M., Ruan J.L., Leboucher S., Dubail M., Heinrich S., Jdey W., Petersson K., Fouillade C., Berthault N., et al. The AsiDNA decoy mimicking DSBs protects the normal tissue from radiation toxicity through a DNA-PK/p53/p21-dependent G1/S arrest. NAR Cancer. 2024;6:zcae011. doi: 10.1093/narcan/zcae011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Berthault N., Bergam P., Pereira F., Girard P.M., Dutreix M. Inhibition of DNA Repair by Inappropriate Activation of ATM, PARP, and DNA-PK with the Drug Agonist AsiDNA. Cells. 2022;11:2149. doi: 10.3390/cells11142149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Lord C.J., Ashworth A. PARP inhibitors: Synthetic lethality in the clinic. Science. 2017;355:1152–1158. doi: 10.1126/science.aam7344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Herath N.I., Berthault N., Thierry S., Jdey W., Lienafa M.C., Bono F., Noguiez-Hellin P., Sun J.S., Dutreix M. Preclinical Studies Comparing Efficacy and Toxicity of DNA Repair Inhibitors, Olaparib, and AsiDNA, in the Treatment of Carboplatin-Resistant Tumors. Front. Oncol. 2019;9:1097. doi: 10.3389/fonc.2019.01097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Le Tourneau C., Delord J.P., Kotecki N., Borcoman E., Gomez-Roca C., Hescot S., Jungels C., Vincent-Salomon A., Cockenpot V., Eberst L., et al. A Phase 1 dose-escalation study to evaluate safety, pharmacokinetics and pharmacodynamics of AsiDNA, a first-in-class DNA repair inhibitor, administered intravenously in patients with advanced solid tumours. Br. J. Cancer. 2020;123:1481–1489. doi: 10.1038/s41416-020-01028-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Chen Y., Li Y., Xiong J., Lan B., Wang X., Liu J., Lin J., Fei Z., Zheng X., Chen C. Role of PRKDC in cancer initiation, progression, and treatment. Cancer Cell Int. 2021;21:563. doi: 10.1186/s12935-021-02229-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Yin X., Liu M., Tian Y., Wang J., Xu Y. Cryo-EM structure of human DNA-PK holoenzyme. Cell Res. 2017;27:1341–1350. doi: 10.1038/cr.2017.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Djuzenova C.S., Fischer T., Katzer A., Sisario D., Korsa T., Steussloff G., Sukhorukov V.L., Flentje M. Opposite effects of the triple target (DNA-PK/PI3K/mTOR) inhibitor PI-103 on the radiation sensitivity of glioblastoma cell lines proficient and deficient in DNA-PKcs. BMC Cancer. 2021;21:1201. doi: 10.1186/s12885-021-08930-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Hafsi H., Dillon M.T., Barker H.E., Kyula J.N., Schick U., Paget J.T., Smith H.G., Pedersen M., McLaughlin M., Harrington K.J. Combined ATR and DNA-PK Inhibition Radiosensitizes Tumor Cells Independently of Their p53 Status. Front. Oncol. 2018;8:245. doi: 10.3389/fonc.2018.00245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Kato T.A., Maeda J., Watanabe H., Kawamura S., Wilson P.F. Simultaneous inhibition of ATM, ATR, and DNA-PK causes synergistic lethality. Biochem. Biophys. Res. Commun. 2024;738:150517. doi: 10.1016/j.bbrc.2024.150517. [DOI] [PubMed] [Google Scholar]
  • 185.Bright S.J., Flint D.B., Martinus D.K.J., Turner B.X., Manandhar M., Ben Kacem M., McFadden C.H., Yap T.A., Shaitelman S.F., Sawakuchi G.O. Targeted Inhibition of DNA-PKcs, ATM, ATR, PARP, and Rad51 Modulate Response to X Rays and Protons. Radiat. Res. 2022;198:336–346. doi: 10.1667/RADE-22-00040.1. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cancers are provided here courtesy of Multidisciplinary Digital Publishing Institute (MDPI)

RESOURCES