Abstract Chemoradiotherapy is the standard of care for many locally advanced cancers, including cervical and head and neck cancers, but many patients cannot tolerate chemotherapy. Clinical trials have shown that radiotherapy combined with hyperthermia (thermoradiotherapy) may be equally effective, yet it yields a suboptimal overall survival of patients, emphasizing the need for improvement. MicroRNAs (miRNAs), short non-coding RNA sequences, are often dysregulated in cancer and exhibit significant potential as radiosensitizers by targeting genes associated with the DNA damage response. In this study, high-throughput miRNA screening of four cervical cancer cell lines identified 55 miRNAs with significant sensitizing potential, with 18 validated across 10 additional cancer cell lines (6 cervical and 4 head and neck). Functional studies of 6 miRNAs, including miR-16, miR-27a, miR-181c, miR-221, miR-224, and miR-1293, showed that they reduced DNA damage repair by downregulating ATM, DNA-PKcs, Ku70/80, and RAD51. Additionally, differential expression of miR-27a, miR-221, and miR-224 in treatment-sensitive versus treatment-resistant patients indicated their predictive biomarker potential for treatment response of cervical cancer patients. Conclusively, this study has identified 18 promising miRNAs for the development of sensitizers for thermoradiotherapy and may provide potential biomarkers for predicting treatment response in locally advanced cancers. Keywords: MT: Non-coding RNAs, miRNA, thermoradiotherapy, DNA damage repair, 3D cell model, locally advanced cancer Graphical abstract graphic file with name fx1.jpg [39]Open in a new tab __________________________________________________________________ Snoek and colleagues identify 18 microRNAs (miRNAs) with significant potential to enhance thermoradiotherapy in cervical and head and neck cancers. By targeting DNA repair pathways, these miRNAs act as radiosensitizers and hold promise for improving therapeutic outcomes. Additionally, miR-27a, miR-221, and miR-224 show potential as biomarkers for predicting treatment response. Introduction Chemoradiotherapy (radiation combined with chemotherapy) is the standard treatment for locally advanced cervical cancer and head and neck cancers. However, many patients cannot tolerate chemotherapy because of frailty and side effects (neurotoxicity, nephrotoxicity, bone marrow depression); thermoradiotherapy (radiation therapy combined with hyperthermia) serves as a well-established alternative treatment strategy.[40]^1 Preclinical and clinical studies have demonstrated a clear benefit of thermoradiotherapy over radiotherapy alone, and non-inferior to chemoradiotherapy with reduced toxicity.[41]^2^,[42]^3^,[43]^4^,[44]^5 The clinical use of hyperthermia as a sensitizer of radiotherapy or chemotherapy is increasingly gaining recognition. Currently, thermoradiotherapy is applied for the treatment of recurrent breast cancer and advanced cervical cancer, and numerous studies have demonstrated the clinical efficacy of thermoradiotherapy for treatment of bladder cancer, esophageal cancer, head and neck cancers, and melanoma.[45]^1^,[46]^6^,[47]^7 Despite these encouraging findings, the clinical outcome of patients with locally advanced cancer remains poor. For locally advanced head and neck cancer patients, the 5-year overall survival rate ranges between 50% and 78%, depending on the specific tumor site.[48]^8 The 5-year overall survival rate for locally advanced cervical cancer patients treated with thermoradiotherapy ranges between 40% and 60%.[49]^9^,[50]^10^,[51]^11 Bladder cancer patients treated with thermoradiotherapy show a reported 3-year overall survival rate of only 28%.[52]^3 These observations underscore the clinical need for improving treatment outcomes. Hyperthermia is employed as a sensitizer of radiotherapy. During hyperthermia, the tumor is locally heated to mild temperatures ranging between 39°C and 42°C for approximately 1 h. This process has been demonstrated to enhance cell sensitivity to radiation through various mechanisms, such as (1) inhibition of radiation-induced DNA damage repair due to temporarily inhibiting the homologous recombination (HR) DNA damage repair pathway, (2) sensitization of S phase cells, and (3) targeting tumor cells in hypoxic and nutrient-deprived areas.[53]^1^,[54]^12^,[55]^13 In addition to its radiosensitizing properties, hyperthermia has been found to trigger systemic anti-tumor immune responses.[56]^14^,[57]^15^,[58]^16 Previously, we have demonstrated that the high specificity of hyperthermia against human papillomavirus (HPV)^+ cervical cancers results from its ability to temporarily restore p53 function by disrupting the binding between the HPV oncoprotein E6 and p53.[59]^17 This p53 restoration leads to induced tumor cell apoptosis following sublethal DNA damage caused by irradiation. In addition, Wang et al. found that hyperthermia leads to the downregulation of HPV oncoprotein E7, effectively reversing HPV-associated carcinogenesis in vitro and in vivo.[60]^18 Furthermore, Jiang et al. showed that phosphorylation of phospholipid scramblase 3, a protein often associated with cancer, is involved in the signal transduction mechanism underlying hyperthermia-induced apoptosis in tongue squamous cell carcinoma.[61]^19 MicroRNAs (miRNAs) are a class of small non-coding RNAs approximately 20–25 nt in length, which play a pivotal role in post-transcriptional gene regulation and are often dysregulated in cancer.[62]^20^,[63]^21 miRNAs have been implicated in tumor radiosensitivity by targeting key components of the DNA damage response.[64]^22 However, most studies have focused on individual miRNAs. No studies have investigated the effects of miRNA modulation on thermoradiotherapy. In the present study, we aim to discover miRNAs that are modifiers of thermoradiotherapy and are potential agents for the development of sensitizers of thermoradiotherapy. Here, we use a high-throughput miRNA screen in three-dimensional (3D) tumor cell spheroids, a model that we recently developed and further optimized to assess the effectiveness of thermoradiotherapy.[65]^23 Comprehensive target prediction, combining computational and experimental approaches, were used to investigate the association of identified miRNAs with DNA damage repair. The function of miRNAs was subsequently studied with clonogenic assays, γ-H2AX foci staining, and automated western blots. The biomarker potential was assessed using miRNA expression profiles of treatment-sensitive and treatment-resistant patients obtained from The Cancer Genome Atlas (TCGA) cervical cancer cohort.[66]^24 Results Discovery screen identifies 55 miRNAs as potent sensitizers for thermoradiotherapy To identify potential sensitizers for thermoradiotherapy, we developed a high-throughput miRNA screening method using 3D spheroid models and cell viability as readout. The discovery screen entailed transfection of cervical cancer cells with 378 miRNA mimics (see [67]materials and methods for selection criteria and [68]Table S1) in 384-well plates followed by irradiation at 2 Gy and hyperthermia at 42°C for 60 min. Three days post-treatment, the cells were assayed for viability. The strategy of this study is outlined in [69]Figure 1A. Figure 1. [70]Figure 1 [71]Open in a new tab Overview of screen strategy and results of the discovery screen across four cervical cancer cell lines (A) Schematic representation of the study outline. (B) Relative effect sizes obtained for each miRNA by comparison to the modal effect (across all miRNAs). miRNAs with a significant treatment effect (FDR <0.1; n = 55) are highlighted in blue. Additionally, miR-140-5p, with a relative effect size of approximately 0.00 is indicated in green. (C) Cell viability results normalized to negative control (NC; gray dotted line) for each cell line (C-4I, CaSki, HeLa, and SiHa), with the modal effect (across all miRNAs; orange dotted line), miR-140-5p as representative “control miRNA,” and the top 5 identified miRNAs indicated. Means ± SDs are shown. Untreated refers to miRNA transfected cells only, while 2 Gy+HT refers to cells that received thermoradiotherapy combined with miRNA transfection. Given that thermoradiotherapy is clinically applied for the treatment of locally advanced cervical cancer, we conducted our discovery screen across four cervical cancer cell lines (C-4 I, CaSki, HeLa, and SiHa). We performed a comprehensive analysis of the obtained data from all cell lines combined to identify miRNAs with a broad sensitizing effect. The miRNA-specific treatment effects, measured by tumor cell viability after treatment, were compared to the modal effect (across all miRNAs) and are indicated as relative effect size, as shown in [72]Figure 1B. Due to our specific focus on the sensitization effect, our statistic model was established to detect a larger treatment effect, resulting in a one-side volcano plot. A false discovery rate (FDR) <0.1 indicates miRNAs with statistically significant treatment effects. Our discovery screen identified 55 miRNAs with significant sensitizing effect to thermoradiotherapy ([73]Figure 1B; [74]Table S1). While the modal effect shows a relative effect size of 0.00, miRNAs with significant sensitizing potential exhibit relative effect sizes ranging between −0.11 and −0.57 ([75]Figure 1B). Consistent effects were observed across all four cervical cancer cell lines ([76]Figure 1C). Notably, C-4 I and HeLa cells were more sensitive to thermoradiotherapy compared to CaSki and SiHa cells. The modal effect observed for each cell line aligns with the cell viability results of the negative control (NC), underscoring the robustness of the statistical model. Validation screen reveals 18 miRNAs with significant sensitizing potential The 55 candidate miRNAs identified in the discovery screen were subsequently validated in an independent set of 10 cancer cell lines, including 6 cervical cancer cell lines (778, 879, C-33A, DoTc2 4510, HT-3, and MS751) and 4 head and neck cancer cell lines (UM-SCC-47, VU-SCC-040, VU-SCC-120, and VU-SCC-147). Head and neck cancer was chosen because of the demonstrated efficacy of hyperthermia in a number of clinical trials.[77]^1^,[78]^25 Additionally, we aimed to explore whether the identified miRNAs exhibit sensitizing effects beyond cervical cancer. Similar as in the discovery screen, we used 3D tumor spheroids for the validation screen and evaluated cell viability 3 days post-treatment (2 Gy irradiation and hyperthermia at 42°C for 60 min). In addition to the 55 candidate miRNAs, we selected 13 miRNAs from the discovery screen as “control” miRNAs, based on their relative effect size of approximately 0.00 ([79]Table S1). These control miRNAs were used to calculate the effect size of the 55 candidate miRNAs. The analysis was conducted on the combined data of all cell lines and identified 18 miRNAs (miR-16, miR-22, miR-27a, miR-92a-1, miR-106b, miR-181a, miR-181c, miR-181d, miR-193a, miR-195, miR-221, miR-224, miR-330, miR-375, miR-455, miR-497, miR-1293, and miR-3158) with a significant sensitizing effect to thermoradiotherapy, demonstrating effect sizes ranging between −0.30 and −1.49 ([80]Figure 2A). Importantly, the thermoradiosensitizing effect mediated by the 18 identified miRNAs demonstrated consistency across most cell lines, as illustrated for the top 5 identified miRNAs in [81]Figure 2B. However, we observed variations in cell line sensitivity to thermoradiotherapy, with cell lines 879, VU-SCC-040 and VU-SCC-120 showing the strongest sensitivity. Notably, miR-224 demonstrated the strongest sensitizing effect in the majority of cell lines. Figure 2. [82]Figure 2 [83]Open in a new tab Results of the validation screen across 10 independent cervical and head and neck cancer cell lines (A) Log2 effect sizes obtained for each miRNA by comparison to the control miRNAs (n = 13), with only significant miRNAs shown (18 out of 55; ∗p < 0.05; ∗∗p < 0.001; ∗∗∗p < 0.00001). The top 5 miRNAs with the strongest sensitizing effect are highlighted in red. (B) Cell viability results for the top 5 identified miRNAs normalized to NCs (controls) on each cell line (untreated as gray dotted lines; 2 Gy+HT as orange dotted lines). Means ± SDs are shown. Statistical analysis of individual cell lines did not reveal significant differences, likely due to the limited number of samples per group (3 vs. 3). Untreated refers to miRNA transfected cells only, while 2 Gy+HT refers to cells that received thermoradiotherapy 1 day after miRNA transfection. Identified miRNAs are associated with DNA repair pathways Previous studies have shown that miRNAs can enhance sensitivity to ionizing radiation by targeting genes involved in DNA damage response.[84]^26 To explore potential gene targets for the 18 miRNAs identified from our screens, we used the ENCORI or miRDB platform for miRNA target prediction,[85]^27 followed by pathway analysis using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) ([86]Figure 3A).[87]^28 Interestingly, pathway enrichment analysis using the predicted targets showed that the ataxia telangiectasia mutated (ATM)-dependent DNA damage response was one of the most significantly enriched pathways for 6 of the 18 miRNAs ([88]Figure 3B; miR-16, miR-181c, miR-193a, miR-224, miR-375, and miR-497), supporting their involvement in inhibiting irradiation-induced double-strain break. For three other miRNAs (miR-27a, miR-330, and miR-455), we also found enrichment of the DNA damage response; however, this was not significant ([89]Figures 3B and [90]S1). Notably, miR-221 showed broad targeting ability on the DNA damage response, as demonstrated by the enrichment of pathways associated with the DNA damage response related to both ATM and ataxia telangiectasia and Rad3-related (ATR) kinases ([91]Figure 3C). An overview of significantly enriched pathways for each miRNA is provided in [92]Figure S1. Most of the predicted miRNA targets were associated with the two main DNA repair pathways: HR and non-homologous end joining (NHEJ) ([93]Table 1). All other predicted targets linked to the DNA damage response are listed in [94]Table S2. Figure 3. [95]Figure 3 [96]Open in a new tab Overview of miRNA target prediction strategy, pathway enrichment, and correlation heatmap (A) Flowchart of miRNA target prediction using the ENCORI (13 miRNAs) or miRDB (5 miRNAs) platform. (B) miRNAs that show significant enrichment for the DNA damage response. Gene ratio indicates the percentage of significant genes over the total genes associated with the DNA damage response. (C) The top 5 enriched pathways for miR-221 predicted targets. (D) Correlation heatmap illustrating miRNA-target co-expression in TCGA cervical cancer dataset for genes associated with DNA repair pathways HR and NHEJ. miRNAs selected for functional validation are indicated in bold. ∗p < 0.05; ∗∗p < 0.01. HITS-CLIP, high-throughput sequencing of RNA isolated by crosslinking immunoprecipitation. Table 1. Overview of predicted miRNA targets associated with DNA damage repair pathways HR and NHEJ miR-16-5p miR-22-5p miR-27a-5p miR-92a-1-5p miR-106b-3p miR-181a-5p miR-181c-5p miR-181d-5p miR-193a-3p miR-195-5p miR-221-3p miR-224-3p miR-330-5p miR-375 miR-455-5p miR-497-5p miR-1293 miR-3158-3p __________________________________________________________________ HR __________________________________________________________________ ATM X X X X X BRCA1 X X MRE11 X X NBN X X X X X PAXIP1 X POLD3 X X X RAD50 X X X X RAD51 X X RAD52 X RAD54B X __________________________________________________________________ NHEJ __________________________________________________________________ LIG4 X NHEJ1 X POLL X POLM X X PRKDC X X X WRN X X XRCC5 X X X X XRCC6 X __________________________________________________________________ Literature on radiosensitizer __________________________________________________________________ yes yes no no no yes no no no yes no no no yes no yes no no CxCa[97]^29 NSCLC[98]^30 PCa[99]^31 HCC[100]^32 CRC[101]^33 CxCa[102]^34 GBM[103]^35 BC[104]^36 CRC[105]^37 LC[106]^38 NSCLC[107]^39 CxCa[108]^41 NB[109]^42 NSCLC[110]^43 OSCC[111]^44 BC[112]^45 CxCa[113]^46 ESCC[114]^47 NSCLC[115]^48 [116]Open in a new tab Predicted targets of each miRNA (n = 18) are marked with an X, along with relevant literature supporting their role as radiosensitizers in cancer. BC, breast cancer; CRC, colorectal cancer; CxCa, cervical cancer; ESCC, esophageal squamous cell carcinoma; GBM, glioblastoma; HCC, hepatocellular cancer; HR, homologous recombination; LC, laryngeal cancer; NB, neuroblastoma; NHEJ, non-homologous end joining; NSCLC, non-small cell lung cancer; OSCC, oral squamous cell carcinoma; PCa, prostate cancer. Given that miRNAs operate by downregulating their gene targets, we computed correlations between the 18 identified miRNAs and genes associated with DNA repair pathways HR and NHEJ using publicly available genome-wide miRNA and messenger RNA (mRNA) expression profiles from cervical and head and neck cancers.[117]^24^,[118]^40 While each miRNA displayed at least one negative correlation, miR-22, miR-27a, miR-193a, miR-221, and miR-455 exhibited negative correlations with the majority of their predicted targets ([119]Figure 3D). Furthermore, a literature search was conducted that revealed supporting evidence for 7 of the 18 identified miRNAs in their role as radiosensitizers in cancer ([120]Table 1). The 6 miRNAs (miR-16,[121]^29^,[122]^30^,[123]^31 miR-22,[124]^32 miR-181a,[125]^33^,[126]^34^,[127]^35 miR-195,[128]^36^,[129]^37^,[130]^38^,[131]^39 miR-375,[132]^41^,[133]^42^,[134]^43^,[135]^44 and miR-497)[136]^45^,[137]^46^,[138]^47^,[139]^48 have been linked to radiosensitivity of cervical cancer and head and neck cancer, but also other cancer types such as breast cancer, colorectal cancer, esophageal and gastric cancer, glioblastoma, hepatocellular cancer, neuroblastoma, lung cancer, prostate cancer, and thyroid cancer ([140]Table 1). Thermoradiotherapy in combination with miRNAs results in reduced DNA damage repair From the 18 identified miRNAs from the validation screen, 6 miRNAs were prioritized for subsequent functional validation. We selected miRNAs with the strongest sensitizing effect observed during the validation screen (effect size of <-0.5; miR-16, miR-27a, miR-181c, miR-224, and miR-1293). Additionally, miR-221 (effect size of −0.38) was included due to its numerous predicted targets associated with DNA repair pathways HR and NHEJ ([141]Table 1). We conducted clonogenic assays in SiHa cells to study cell survival following different treatment combinations, including miRNA mimic alone (untreated), irradiation (2 Gy) combined with miRNA mimics, and thermoradiotherapy (2 Gy and 42°C for 60 min) combined with miRNA mimics. We observed a significantly lower survival fraction when thermoradiotherapy was combined with miR-221, miR-224, or miR-1293 mimics compared to thermoradiotherapy combined with a NC ([142]Figure 4A; representative images of the colony-forming assay are included in [143]Figure S2). Upregulated expression of miRNA after transfection was measured using quantitative reverse-transcription PCR (RT-qPCR) ([144]Figure S3B), with a fold change relative to the NC ranging from 10^2 to 10^8. Figure 4. [145]Figure 4 [146]Open in a new tab Response to irradiation (2 Gy), thermoradiotherapy (2 Gy+HT), and miRNA mimics in SiHa cells (A) Clonogenic survival fractions for each miRNA are shown and compared to NC. (B) Representative images of y-H2AX foci staining (in green) for each condition. (C) The number of y-H2AX foci (the number of double-stranded breaks) 24 h after thermoradiotherapy combined with miRNA mimics or NC. Each graph represents the γ-H2AX foci in each cell, with a minimum of 70 cells per experiment. (D) Normalized protein expression against NC. Means ± SDs are shown. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001 Furthermore, we investigated residual (unrepaired) DNA double-stranded breaks (DSBs) 24 h post-treatment using γ-H2AX foci staining. Thermoradiotherapy combined with either miR-16, miR-27a, miR-181c, miR-221, miR-224, or miR-1293 demonstrated a significant increase in DNA DSBs compared to the control, with the most pronounced increase observed for miR-221 ([147]Figures 4B and 4C). We subsequently performed western blot analysis to study the impact of miRNA modulation on the protein expression of several predicted targets associated with DNA repair pathways, specifically HR and NHEJ ([148]Figures 4D and [149]S4). Transfection of SiHa cells with miRNA mimics miR-16, miR-27a, miR-181c, miR-221, miR-224, or miR-1293 led to a reduction in ATM, a key player in HR. Regarding the regulation of NHEJ, we found strong effects for all six miRNAs on the protein expression of DNA-dependent protein kinase catalytic subunit (DNA-PKcs). In addition, we analyzed the protein expression of two other key players in NHEJ: Ku70 (gene name XRCC6) and Ku80 (gene name XRCC5). We observed that miR-1293 strongly reduced the protein expression of Ku80, while Ku70 expression was affected by miR-27a, miR-221, and miR-224. Additionally, these three miRNAs also reduced the expression of RAD51, a pivotal player in HR. miR-27a, miR-221, and miR-224 exhibit predictive biomarker potential for treatment response Next, we explored the predictive biomarker potential of the identified miRNAs. To accomplish this, we utilized genome-wide miRNA expression profiles from advanced stage cervical cancer patients obtained from TCGA.[150]^24 Unfortunately, TCGA dataset lacks information on cervical cancer patients who have undergone thermoradiotherapy, requiring us to focus on chemoradiotherapy instead. Using the clinical data from TCGA, we defined two groups of cancer patients based on their response to treatment: treatment-sensitive (n = 37), including those with complete and partial responses, and treatment-resistant (n = 18), including those with stable and progressive disease. Interestingly, we found that miR-27a, miR-221, and miR-224 are significantly downregulated in treatment-resistant cervical cancer patients ([151]Figure 5), indicating their potential as predictive biomarkers for treatment response in cervical cancer patients. Figure 5. [152]Figure 5 [153]Open in a new tab Expression profiles of miR-16, miR-27a, miR-181c, miR-221, and miR-224 in TCGA cervical cancer dataset Cancer patients were divided into two groups based on their response to chemoradiotherapy: treatment sensitive (n = 37), including those with complete and partial responses, and treatment resistant (n = 18), including those with stable and progressive disease. ∗p < 0.05; ∗∗p < 0.01. RPM, reads per million. Discussion Chemotherapy-based radiosensitization is the standard of care for treating locally advanced cervical and head and neck cancers. However, many patients cannot tolerate chemotherapy, and clinical trials have shown that thermoradiotherapy may be an equally effective alternative. miRNAs have previously been studied as potential targets for radiosensitization, but none of these studies have studied their potential role in thermoradiotherapy sensitization. In the present study, we identified and validated 18 miRNAs with significant sensitizing potential for thermoradiotherapy in 14 cervical and head and neck cancer cell lines. Functional validation showed that miRNA-mediated sensitization was achieved by targeting genes associated with DNA repair pathways, including RAD51 (HR) and Ku80 (NHEJ). An important aspect of this study is that we used a high-throughput miRNA screen on 3D spheroid models with cell viability as the readout, rather than conventional clonogenic assays to assess cell survival after treatment. Our approach overcomes the practical limitations of the clonogenic assay, including the long experiment duration, low sample throughput and time-consuming colony counting. The validity of our approach is supported by several findings. First, using the cellular ATP content as a measure of cell viability, we found a similar variation in sensitivity among cervical cancer cell lines and head and neck cell lines as reported by others using clonogenic assays.[154]^49 This underscores the reliability of exploring treatment sensitization by measuring cell viability in a 3D model, while overcoming the difficulty when certain cell lines do not form colonies. Second, we observed similar sensitizing effects for most miRNAs when comparing results from clonogenic survival assays with cell viability results ([155]Figure 4B). This is consistent with previous studies showing similar results between these two types of assays.[156]^50^,[157]^51 Third, among the discovered thermoradiosensitizing miRNAs, we identified several miRNAs previously described as potent radiosensitizers in cancer, including miR-16-5p, miR-181a-5p, miR-195-5p, miR-221-3p, miR-375-3p, and miR-497-5p ([158]Table 1). Notably, several of these miRNAs have been described as radiosensitizers in cervical and/or head and neck cancers. For example, miR-16-5p has been shown to modulate the radiosensitivity of cervical cancer cells by regulating coactivator-associated arginine methyltransferase 1 (CARM1).[159]^29 CARM1 plays a critical role in the DNA damage response by methylating p300, which enhances the interaction between p300 and BRCA1 (key player in HR), thereby promoting BRCA1 transcriptional activity.[160]^52 Furthermore, miR-181a-5p was found to target Raf kinase inhibitor protein in nasopharyngeal cancer cells thereby enhancing radiotherapy resistance.[161]^53 In addition, Ke and colleagues found that miR-181a-5p can confer radiotherapy resistance in cervical cancer cells by targeting pro-apoptotic protein kinase C delta (PRKCD) and that its expression may serve as a prognostic biomarker for radiotherapy response.[162]^34 On top we observed mostly consistent sensitizing effects in different cancer types (cervical and head and neck cancer), highlighting the potential broad applicability of these miRNAs in improving therapeutic outcomes. Some observed differences between cell lines are most likely due to inherent differences in radiation sensitivity and/or differences in endogenous miRNA levels between cell lines ([163]Figure S3A).[164]^54 Interestingly, Western blot analysis revealed downregulation of ATM and DNA-PKcs (gene name PRKDC) after transfection of all six miRNAs ([165]Figure 4D). As early sensors of DNA DSBs, ATM and DNA-PKcs activate a series of kinase reactions essential for DNA DSB repair.[166]^55^,[167]^56 Therefore, the downregulation of ATM and DNA-PKcs could inhibit DNA damage repair, which is consistent with our findings from γ-H2AX stainings. However, the types of interactions may vary among the miRNAs. The negative correlation in expression levels between three miRNAs (miR-27a, miR-181c, and miR-221) and ATM was confirmed by TCGA data ([168]Figure 3D). Target prediction using a combination of computational and experimental strategies (high-throughput sequencing of RNA isolated by crosslinking immunoprecipitation [HITS-CLIP] data) also supports the interaction between these miRNAs and their targets, suggesting that ATM is directly targeted by miR-27a, miR-181c, and miR-221. For DNA-PKcs, although TCGA data show a negative correlation between the miRNAs and PRKDC, target prediction did not provide evidence for a direct interaction between the miRNAs and PRKDC, suggesting an indirect effect of the miRNAs. Repair of radiation-induced DNA DSBs is typically accomplished by two pathways: NHEJ and HR.[169]^57 During classical NHEJ, DNA DSBs are initially recognized by the Ku70-Ku80 heterodimers, which recruit and promote the binding of other NHEJ proteins, including DNA-PKcs, ligase 4, and XRCC4.[170]^58 Thus, downregulation of Ku70 (observed with miR-16, miR-181c, miR-221, miR-224, and miR-1293) and Ku80 (observed with miR-16, miR-27a, miR-221, miR-224, and miR-1293) could inhibit NHEJ. While miR-16[171]^29 and miR-1293[172]^59 have been reported to target DNA repair genes, their effects on Ku70 and Ku80 have not been previously demonstrated. Target prediction supports miR-16 targeting of Ku80 (XRCC5), but the mechanism by which miR-1293 inhibits DNA damage repair requires further investigation. Alternatively, DNA DSBs can also be repaired by HR. Following initiation resection by the MRN complex (MRE11-RAD51-NBS1) and activation of ATM, HR is catalyzed by several enzymes, in particular by recombinase RAD51.[173]^60 RAD51 plays an important role in HR by assembling filament on DNA, interacting with other proteins, and then facilitating HR.[174]^61 In our validation, miR-221 mimic transfection resulted in a pronounced downregulation of RAD51 protein levels. This observation is further supported by computational and experimental target prediction, suggesting that miR-221 may inhibit HR by directly affecting RAD51 levels ([175]Figure 3D). These results are summarized in a network model shown in [176]Figure 6, which illustrates our proposed mechanism of miRNA-mediated sensitization of thermoradiotherapy. Figure 6. [177]Figure 6 [178]Open in a new tab Network model illustrating our proposed mechanism of miRNA-mediated sensitization to thermoradiotherapy based on the results of this study Direct regulation is indicated by black solid line, and indirect regulation is indicated by gray dotted line. One limitation of our study is the use of single-dose irradiation (2 Gy) instead of applying a clinically relevant treatment regimen. For locally advanced cervical cancer, the clinical treatment regimen typically involves fractionated radiotherapy with 20–25 fractions and a total dose of 45–50 Gy.[179]^10 However, single-dose irradiation is often used in in vitro studies to simplify experimental conditions and allow for controlled analysis of cellular responses to radiation.[180]^49 Future studies using fractionated radiation may provide more clinically relevant insights and potentially confirm our findings. Additionally, our functional validation was conducted exclusively in cervical cancer cell lines. Expanding validation to other cancer cell lines, particularly head and neck cancer cells, as well as non-malignant cells, may strengthen our findings and provide valuable insights into the toxicity profile of the miRNAs. miRNA toxicity is a critical issue that could significantly impact therapeutic efficiency, making it essential to evaluate the toxicity profile of each miRNA. In this study, we used a 2-nM miRNA mimic concentration for each cell line, which was selected based on prior optimization efforts.[181]^23^,[182]^62 While miR-224 exhibited the strongest sensitizing effects across cell lines, it also notably reduced cell viability before treatment ([183]Figure 2B), suggesting general toxicity. To address this, we tested a lower concentration of miR-224 and found that it reduced its impact on cell viability ([184]Figure S5), particularly in HeLa and SiHa cells, demonstrating a dose-dependent effect. A similar trend was observed for miR-16, miR-27a, and miR-1293. These findings highlight the need for further optimization of miRNA mimic concentrations in future studies to minimize potential toxicity while maintaining therapeutic efficacy. While this study primarily focused on miRNA-mediated modulation of the two main DNA repair pathways, HR and NHEJ, other DNA damage response-associated genes remain largely unexplored in this context. Additionally, our pathway analysis revealed several other significantly enriched pathways, including those related to angiogenesis, apoptosis, cell-cycle regulation and immune responses, as shown in [185]Figure S1. Further investigation into these pathways and other DNA damage response-associated genes may provide additional leads for therapeutic intervention and could further advance our understanding of miRNA-mediated treatment sensitization. The field of miRNA-based therapeutics has gained significant interest due to the central role of miRNAs in gene regulation and their involvement in various cancers. Several miRNA mimics and inhibitors, such as miR-10b, miR-16, miR-34a, miR-155, and miR-193a, have been or are currently being investigated in clinical trials for the treatment of cancer and other diseases.[186]^63^,[187]^64 Unlike single-gene therapies, miRNA-based therapeutics offer a unique advantage due to their ability to simultaneously target multiple pathways (i.e., HR and NHEJ). However, despite promising preclinical results, the translation of miRNA therapeutics into clinical practice remains highly challenging.[188]^65 One major hurdle of miRNA therapeutics is the difficulty in achieving efficient and specific delivery. miRNA mimics and inhibitors require stable delivery systems to prevent degradation and ensure targeted cellular uptake, yet existing strategies often lead to off-target effects, immune activation, or insufficient bioavailability. Early clinical trials, such as a phase 1 trial for MRX34 (a miR-34a mimic; this study was registered at [189]ClinicalTrials.gov: [190]NCT01829971),[191]^66 a phase 2 trial for lademirsen (a miR-21 inhibitor; this study was registered at [192]ClinicalTrials.gov: [193]NCT02855268),[194]^67 and phase 2 trials for Miravirsen (a miR-122 inhibitor; these studies were registered at [195]ClinicalTrials.gov: [196]NCT01872936 and [197]NCT02031133)[198]^68 encountered setbacks due to severe immune-mediated toxicities or limited efficacy, ultimately leading to trial discontinuation. These examples highlight the complexities of miRNA-based therapeutics, particularly regarding safety and specificity. Nanoparticle-based delivery systems have shown promise in overcoming some of these challenges by enhancing stability and targeting efficiency.[199]^64 In the context of cervical cancer, localized delivery approaches could mitigate systemic toxicity.[200]^69 However, while the anatomical accessibility of the cervix may facilitate localized administration, this does not inherently resolve broader delivery challenges, including endosomal sequestration and biodistribution concerns. Furthermore, tumor heterogeneity, hypoxic environments, and variable miRNA expression profiles within cervical cancer further complicate therapeutic efficacy and patient stratification. Despite these challenges, miRNA therapeutics continue to hold potential, particularly when integrated into multimodal treatment strategies. For example, in thermoradiotherapy, hyperthermia enhances tumor vasculature permeability, which could improve nanoparticle uptake and facilitate miRNA-based radiosensitization.[201]^70 However, preclinical and clinical studies are needed to determine the safety and efficacy of such approaches. In addition to their role as therapeutic agents, this study has identified several miRNAs that show potential as predictors of treatment response in cervical cancer patients. Specifically, miR-27a, miR-221, and miR-224 show significant downregulation in treatment-resistant patients, suggesting their utility as predictive markers of treatment response. In support of these findings, Ren and colleagues demonstrated that miR-27a plays a critical role in modulating the radiosensitivity of triple-negative breast cancer cells, suggesting that miR-27a expression could serve as a valuable marker to identify patients who will benefit from radiotherapy treatment.[202]^71 In addition, Upraity et al. found that increased miR-224 expression increased the radiosensitivity of glioblastoma cells.[203]^72 Notably, TCGA cervical cancer dataset used in our study only includes patients treated with radiotherapy or chemoradiotherapy. While our study focuses on thermoradiotherapy, the potential biomarker utility of the identified miRNAs in the context of chemoradiotherapy underscores their broad applicability. This suggests that our findings could be extrapolated to other combined-modality treatments that include radiotherapy. Further validation studies in larger patient cohorts and in other cancer types are warranted to confirm the predictive value of miR-27a, miR-221, and miR-224 and to explore their applicability in clinical practice. In conclusion, this study highlights the potential role of miRNAs as sensitizers of thermoradiotherapy and improves our understanding of the molecular mechanisms underlying miRNA-mediated sensitization of thermoradiotherapy. Further research on the clinical translation of these findings, including prospective validation studies and mechanistic investigations, is warranted to fully exploit the therapeutic potential of miRNA-based approaches in combination with thermoradiotherapy for the treatment of locally advanced solid tumors. Materials and methods Cell lines and cell culture Human cervical cancer cell lines C-33A, C-4 I, CaSki, DoTc2 4510, HeLa, MS751, and SiHa were obtained from the American Type Culture Collection (ATCC, Manassas, VA) and cultured in Dulbecco’s modified Eagle’s medium (DMEM) formulated with high glucose and l-glutamine (Thermo Fisher Scientific, Waltham, MA) and supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin, and 100 μg/mL streptomycin (all from Thermo Fisher Scientific). Human cervical cancer cell line HT-3 was obtained from the ATCC and cultured in McCoy’s 5a medium supplemented with 10% FBS, 100 U/mL penicillin, and 100 μg/mL streptomycin (all from Thermo Fisher Scientific). Two cervical cancer cell lines, 778 and 879, were previously provided by Dr. P.L. Stern (Paterson Institute for Cancer Research, Manchester, UK) and cultured in RPMI 1640 medium (Thermo Fisher Scientific) supplemented with 10% FBS, penicillin at 100 U/mL, and streptomycin at 100 μg/mL (all from Thermo Fisher Scientific).[204]^73 Head and neck squamous cell carcinoma (HNSCC) cell lines UM-SCC-47, VU-SCC-040, VU-SCC-120, and VU-SCC-147 were obtained as described previously and cultured with DMEM supplemented with 10% FBS, 1 mM sodium pyruvate, 100 U/mL penicillin, and 100 μg/mL streptomycin (all from Thermo Fisher Scientific).[205]^74^,[206]^75^,[207]^76 Cell cultures were maintained at 37°C in a humidified atmosphere with 5% CO[2]. Cell lines were authenticated by short-tandem repeat (STR) testing, monthly tested for the presence of mycoplasma and HPV status was confirmed using the QIAscreen HPV PCR assay (Qiagen, Hilden, Germany). Irradiation and hyperthermia Cells were irradiated with 2 Gy of γ-radiation at room temperature using the Gammacell 220 Cobalt irradiator with a ^131Cs source (MDS Nordion, Ottawa, Canada). Hyperthermia treatment of cells started within 30 min after irradiation. Incubation of cells was performed by partially submerging the plates in a thermostatically controlled water bath (VWR International, Amsterdam, the Netherlands) for 60 min at 42°C (±0.1°C). The temperature was measured in parallel plates with a thermometer, and the desired temperature was reached in approximately 8 min for 384-well plates and 5 min for 6-well plates. High-throughput miRNA screens For the discovery screen, four cervical cancer cell lines (C-4 I, CaSki, HeLa, and SiHa) were subjected to high-throughput reverse transfection using 384-well ultra-low attachment plates (Corning, New York, NY) to allow for 3D cell cultures (spheroids). A total of 378 miRNAs ([208]Table S1) were selected based on in-house and publicly available genome-wide miRNA expression data from cancer types relevant to our study (i.e., cervical cancer, head and neck cancers) as well as literature supporting their role as a radiosensitizer in cancer.[209]^22^,[210]^24^,[211]^40^,[212]^77^,[213]^78 Cells were transfected using an optimized reverse transfection protocol, in which efficiency was carefully assessed through miRNA qPCR, miRNA target western blot analysis, and cellular viability measurements, as described previously.[214]^23^,[215]^62 In brief, on day 0 transfection complexes were prepared by mixing 0.04 μL DharmaFECT4 transfection reagent (Horizon Discoveries, Cambridge, UK) and 2 nM (end concentration) of miRNA mimic (miRIDIAN miRNA mimic library version 19 [Horizon Discoveries]) with 20 μL Opti-MEM serum-free medium (Thermo Fisher Scientific). Cells were harvested, resuspended in culture medium, and plated in a 384-well format (500 cells/20 μL/well) simultaneously with the addition of transfection complexes. Plates were sealed with breathable film to prevent evaporation. On day 1, cells were treated with thermoradiotherapy or kept in culture as a control. Cell viability was assessed on day 4 by adding 15 μL Cell-Titer Glo 3D (Promega, Madison, WI) per well, after which luminescence was measured using an Infinite F200 microplate reader (Tecan Group, Männedorf, Switzerland). The discovery screen was performed in triplicate. For the validation screen, 10 additional cancer cell lines were utilized, including 6 cervical cancer cell lines (778, 879, C-33A, DoTc2 4510, HT-3, and MS751) and 4 HNSCC cell lines (UM-SCC-47, VU-SCC-040, VU-SCC-120, and VU-SCC-147). From the discovery screen, 55 candidate miRNAs along with 13 control miRNAs were selected for validation. The setup and readout of the validation screen was similar to that of the discovery screen, including the amount of cells, transfection reagent, and miRNA mimic concentration. The validation screen was performed in triplicate. miRNA transfection for functional assays SiHa cells were reverse transfected with miRNA mimics for hsa-miR-16-5p, hsa-miR-27a-5p, hsa-miR-181c-5p, hsa-miR-221-3p, hsa-miR-224-3p, hsa-miR-330-5p, hsa-miR-1293, or NC #2 (C-300483-03, C-301028-01, C-300556-03, C-300578-05, C-301304-00, C-301082-01, C-301347-00, CN-002000-01 [Horizon Discoveries]) in ultra-low attachment plates following the same protocol outlined for the miRNA screens. In brief, transfection complexes were prepared by mixing 2 μL (end volume 0.04 μL) DharmaFECT4 transfection reagent (Horizon Discoveries) and 2 nM (end concentration) of miRNA or control mimic with 200 μL Opti-MEM serum-free medium (Thermo Fisher Scientific). Harvested cells were plated in 6-well ultra-low attachment plates (Corning) at a density of 85,000 cells/1.8 mL/well. RNA isolation Total RNA was isolated from cells using the miRNeasy Tissue/Cells Advanced kit, including on-column DNase treatment, according to the manufacturer’s instructions (Qiagen). RNA concentrations were measured using NanoDrop One (Thermo Fisher Scientific). RT-qPCR This study was performed in compliance with the minimum information for publication of qPCR experiments guidelines, where applicable.[216]^79 Reverse transcription was performed using the TaqMan microRNA Reverse Transcription kit (4366596; Thermo Fisher Scientific). The manufacturer’s protocol was adapted to multiplex reverse transcription for all miRNAs and the reference gene, as described previously.[217]^77^,[218]^80 In brief, 50 ng total RNA was reverse transcribed in 16.5-μL reactions containing 9 μL primer pool, 0.45 μL deoxynucleotide triphosphate (100 mM), 2.25 μL 10× RT buffer, 0.3 μL RNase inhibitor (20 U/μL), and 4.5 μL MultiScribe Reverse Transcriptase. Expression of hsa-miR-16-5p, hsa-miR-27a-5p, hsa-miR-181c-5p, hsa-miR-221-3p, hsa-miR-224-3p, hsa-miR-330-5p, and hsa-miR-1293 was determined using TaqMan microRNA assays (000391, 002445, 000482, 000542, 121210_mat, 002230, 002905; Thermo Fisher Scientific). U75 was included as a reference gene (001219; Thermo Fisher Scientific). qPCR was performed in 10-μL reactions containing 5 μL TaqMan Universal Master Mix II, no UNG (4440043; Thermo Fisher Scientific), 0.5 μL miRNA-specific TaqMan microRNA assays (catalog numbers for each miRNA assay are listed above), 3.5 μL H[2]O, and 1 μL cDNA. qPCR reactions were performed on the ABI 7500 Fast Real-Time PCR System (4351107; Thermo Fisher Scientific) using the following conditions; 5 min at 95°C followed by 40 cycles of 15 s at 95°C and 1 min at 60°C. H[2]O was tested as a negative PCR control. qPCR analysis and determination of Ct values were conducted using Applied Biosystem 7500 Software version 2.3 (4397808; Thermo Fisher Scientific). The miRNA expression levels were normalized to U75 by applying the 2^−ΔCt method.[219]^81 Clonogenic assay Clonogenic assays were performed as described previously.[220]^82^,[221]^83 Cells were collected 2 days post-miRNA transfection (to harvest enough cells for the subsequent protocol) and plated at different densities depending on the treatment group (50, 100, 200, 500, 1,000 or 2,000 cells/6-well) 4–8 h prior to treatment. Subsequently, cells were treated with either irradiation (2 Gy) alone or thermoradiotherapy (2 Gy and 42°C for 60 min) and incubated for 10–12 days to allow for colony formation. The plates were stained with a methanol-crystal violet solution (5 mg/mL crystal violet in 10% ethanol, 50% methanol, and 40% water) followed by scanning and counting with GelCount (Oxford Optronix, Abington, UK). Plating efficiencies were defined by dividing the amount of counted colonies by the amount of seeded cells. Survival fractions were calculated by normalization to the plating efficiency of untreated cells. γ-H2AX immunofluorescence assay SiHa cells were reverse transfected in the 8-well Nunc Lab-Tek II Chamber Slide system (Thermo Fisher Scientific) by mixing 0.6 μL (end volume 0.04 μL) DharmaFECT4 transfection reagent (Horizon Discoveries) and 2 nM miRNA or control mimic with 80 μL opti-MEM serum-free medium (Thermo Fisher Scientific). Harvested cells were seeded at 9,000 cells/520 μL/well. Subsequently, cells were treated with ionizing radiation (2 Gy) and hyperthermia (42°C for 60 min) and 24 h post-treatment the cells were fixed with 2% paraformaldehyde and permeabilized with 0.1% Triton X-100 in phosphate-buffered saline (PBS). Slides were then incubated with 1:200 Alexa Fluor 488 anti-H2A.X Phospho (Ser139) antibody (catalog no. 613405; BioLegend, San Diego, CA) and counterstained with 1 μg/mL DAPI. Slides were then visualized with a Nikon A1R HD25 confocal microscope (Nikon Europe, Amstelveen, the Netherlands). H2AX foci numbers were counted manually and at least 70 cells per condition per experiment were scored in three independent experiments. Automated western blot SiHa cells were collected 24 h post-miRNA transfection and plated in 6-well plates at a density of 85,000 cells/well 4–8 h prior to treatment. Cells were treated with irradiation (2 Gy) and hyperthermia (42°C for 60 min), and 4 h post-treatment cells were harvested. Whole-cell lysates were prepared on ice using the RIPA Lysis Buffer System (Santa Cruz Biotechnology, Dallas, TX), according to the manufacturer’s instructions. Protein concentrations were determined using Pierce bicinchoninic acid protein assay following the manufacturer’s instructions. The automated western blots were conducted using JESS Simple Western instrument (ProteinSimple, Bio-Techne, Minneapolis, MN). Proteins (3 μg) were diluted in 0.1× sample buffer and 1× fluorescent master mix (EZ standard pack; Bio-Techne) to a total volume of 6 μL/well and separated using a 12- to 230-kDa or a 66- to 440-kDa separation module (Bio-Techne). Primary antibodies were diluted in antibody diluent 2 (Bio-Techne) and included rabbit anti-ATM (1:50, catalog no. 2873; Cell Signaling Technologies, Danvers, MA), mouse anti-β-actin (1:12.5, catalog no. 3700s; Cell Signaling Technologies), rabbit anti-DNA-PKcs (1:100, bsm-52493R, Bioss, Woburn, MA), rabbit anti-Ku70 (1:500, catalog no. PA5-27538, Thermo Fisher Scientific), rabbit anti-Ku70/Ku80 (1:500, catalog no. PA-5-27538; Thermo Fisher Scientific), and mouse anti-RAD51 (1:50, catalog no. MS-988-P1; Thermo Fisher Scientific). Secondary antibody detection modules anti-mouse and anti-rabbit were used according to the manufacturer’s instructions. The RePlex module (Bio-Techne) was used to allow sequential detection of multiple proteins. Data were analyzed using the software Compass for Simple Western version 6.3. miRNA target prediction Target prediction of selected miRNAs from the validation screen (n = 18) was performed using the ENCORI platform version 2.0[222]^27 or miRDB ([223]https://mirdb.org).[224]^84 The ENCORI platform was employed for 13 miRNAs (miR-16-5p, miR-106b-5p, miR-181a-5p, miR-181c-5p, miR-181d-5p, miR-193a-5p, miR-195-5p, miR-221-3p, miR-224-3p, miR-330-5p, miR-375, miR-455-5p, and miR-497-5p). No predicted targets were identified for 5 miRNAs (miR-22-5p, miR-27a-5p, miR-92a-1-5p, miR-1293, and miR-3158-3p) using the ENCORI platform, leading us to utilize miRDB for target prediction. Targets predicted by ENCORI with a HITS-CLIP number ≥4 and targets by miRDB with a prediction score ≥60 were subjected to pathway analysis using DAVID version 2024q2,[225]^28 with WikiPathways version 20241110 as reference. Pathways associated with the DNA damage response were selected for further investigation. External expression data of cervical tissue specimens Genome-wide miRNA and mRNA expression profiles along with clinical data from cervical cancer patients[226]^24 and head and neck cancer patients[227]^40 was obtained from TCGA. Pearson correlation coefficients (r) were computed between miRNAs and their predicted miRNA targets and visualized in a correlation heatmap. Patients with advanced cervical cancer were classified into two groups according to their response after chemoradiotherapy: treatment sensitive (n = 37), including complete and partial response, or treatment resistant (n = 18), including stable and progressive disease. Statistical analysis Analysis of high-throughput screens As the number of cell lines was relatively small in the discovery screen, we opted for a simple analysis model per miRNA: a linear mixed effect model for the log[2](response), with thermoradiotherapy treatment and cell lines as fixed factors and assay (biological replicates) as a random effect.[228]^85 miRNA-specific treatment effects were contrasted with the modal effect (across all miRNAs) to filter out effects that are mostly due to the treatment alone. Hence, resulting miRNAs have, in conjunction with the treatment, a significantly larger effect than the modal effect. An FDR of ≤0.10 was used as threshold for significance. The discovery screen data was analyzed using the R-package ShrinkBayes version 2.13.7, which enables efficient estimation for large screens and small sample sizes.[229]^85 From the results of the discovery screen, 13 miRNAs were designated as control miRNAs based on their relative effect size (around 0.0; [230]Table S2). For the validation screen, the utilization of a larger number of cell lines allowed for a slightly more complex model, also to disentangle the treatment and miRNA effect. For each miRNA, a linear mixed-effects model was used on the log[2](response) using miRNA presence (yes/no), thermoradiotherapy treatment (yes/no), cancer type (cervical cancer/HNSCC), and HPV status (negative/positive) as fixed effects (the latter two as potential confounders), plus cell line, assay (biological replicates nested within cell line), and pair (nested within assay) as random effects. The first accounts for variation across cell lines (now modeled as random effects as we have 11 levels), the second for correlation between technical repeats of screens, and the last accounts for correlation between responses of the parental (no miRNA) and non-parental settings within one screen. This model was fit for both control miRNAs and the candidate miRNAs were corrected for the mean effect of the control miRNAs (by subtracting the latter from the responses of the candidate miRNAs) to ensure that selected miRNAs have significantly stronger effects than the control miRNAs. Significance of the miRNA was tested using a likelihood ratio test (chi-squared; df = 1) of the model against the null model, which contains all factors except for the miRNA presence covariate. Likewise, treatment-miRNA interactions were tested by adding such an interaction to the model and testing it against the model with main effects only. Resulting p values were Bonferroni corrected for multiple testing, and effect size estimates were back-transformed to the original scale, rendering fold changes. The R-package lme4 version 1.1–35 was used to fit the mixed models.[231]^86 Analysis of functional assays All data are expressed as mean ± SD. The statistical significance of clonogenic assays, γ-H2AX immunofluorescence assays, and immunoblotting was tested using the Kruskal-Wallis test with the post hoc Dunn’s test for comparison to NC within the same treatment group (untreated, 2 Gy or 2 Gy+HT). Comparison of miRNA expression profiles between radio-sensitive and radio-resistant groups was made using the Mann-Whitney test (∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001). Correlation between miRNA and mRNA expression was tested using Spearman correlation (∗p < 0.05; ∗∗p < 0.01). Analyses of functional experiments were performed with GraphPad Prism version 10 (GraphPad Software, La Jolla, CA). Statistical analyses of publicly available data were performed with R (version 4.2.1, [232]https://www.R-project.org/). Data processing and figures were generated using the readr (version 2.1.5), dplyr (version 1.1.4) and ggplot2 (version 3.4.4) packages. Study outline and network model were illustrated using BioRender ([233]https://www.biorender.com/) and Adobe Illustrator 2023. Data availability mRNA and miRNA expressions and treatment response information are available from TCGA dataset. All other data supporting the findings of this study are available from the corresponding author upon reasonable request. Acknowledgments