3.1. Non-coding RNAs and sorafenib combination improves the therapeutic response
Non-coding RNAs are often deregulated in HCC and are extensively involved in the modulation of molecular mechanisms leading to sorafenib resistance, such as hypoxia, autophagy, metabolic reprogramming, and activation of oncogenic pathways [
12]. Sorafenib is an oral TKI that blocks tumor cell proliferation by targeting Raf/MEK/ERK signaling at the level of Raf kinase, and exerts an antiangiogenic effect by targeting vascular endothelial growth factor receptor-2/-3 (VEGFR-2/-3), and platelet derived growth factor receptor beta (PDGFR-β) tyrosine kinases [
43].
Due to the antiangiogenic properties of sorafenib, blocking factors having a mitogenic effect on endothelial cells and interfering with HIF-1A signaling represent crucial actions to potentiate sorafenib efficacy and to prevent drug resistance. MicroRNA-494 is an oncogenic HCC-associated miRNA which is upregulated in 30% of cases and associates with stem cell-like characteristics and poor prognosis [
44,
45]. Regarding its involvement in sorafenib resistance, we previously demonstrated that miR-494 activates the AKT/mTOR pathway by targeting PTEN and reported a stronger antitumor effect of antagomiR-494 plus sorafenib treatment with respect to sorafenib alone in the DEN-HCC rat model [
46]. Notably, the Golgi phosphoprotein GOLPH3 is involved in sorafenib resistance in vivo by increasing the microvascular density of xenograft tumors. Gao et al. showed that exosomes released by GOLPH3 overexpressing HCC cells are enriched in miR-494 content enhancing tube formation and migration of the umbilical endothelial HUVEC cell line. MiR-494-loaded extracellular vesicles increased sorafenib resistance of HCC cells, highlighting both autologous and heterologous mechanisms of action for this miRNA [
47]. These data proved the biologic activity of exosome-associated miR-494 in cell-to-cell crosstalk and confirmed that miR-494 as an important tumor-derived autocrine and paracrine signal promoting angiogenesis, HIF-1A activation, and tumor growth under hypoxic condition in different cancer types [
48]. Similarly, the polypeptide 14-3-3η is a growth-promoting factor highly expressed in tumor and vascular endothelial cells contributing to poor survival of HCC patients [
49]. The study by Shen and coworkers described the overexpression of 14-3-3η in sorafenib-resistant (SR) Huh-7 cells and demonstrated that its silencing restores drug sensitivity and reduces cancer stem cell (CSC) properties. Interestingly, 14-3-3η polypeptide post-transcriptionally activates HIF-1A via inhibition of the proteasome machinery. MiR-16 was identified as the epigenetic regulator of this polypeptide showing an inverse correlation in HCC patients treated with combined transartherial chemoembolization (TACE) and sorafenib. In line, low miR-16/high 14-3-3η HCC subgroup showed the worst OS after combined treatment. MiR-16 overexpression or 14-3-3η silencing in combination with sorafenib determined a higher anti-tumor effect in xenograft mice with respect to sorafenib alone, highlighting miR-16 restoration as a promising strategy to improve sorafenib efficacy in HCC [
50]. A genome-wide CRISPR/Cas9 library screening identified the deficiency of miR-15a (belonging to the tumor suppressor miR-15a/16-1 miRNA cluster) and miR-20b (belonging to the oncogenic miR-19~92 miRNA cluster) to contribute to sorafenib resistance in HCCLM3 cell line. In agreement with the opposite role of these two miRNA clusters in tumors, miR-15a overexpressing cells decreased in vivo tumorigenesis while miR-20b overexpression slightly increased tumor size; nevertheless, the overexpression of both miRNAs led to inhibition of tumorigenesis in the xenograft model subjected to sorafenib treatment, confirming their role in drug sensitization. Target prediction algorithms identified the cochaperone CDC37L1 as the only common target of these two miRNAs. Functional analysis and luciferase assays proved its inhibition by both miR-15a and miR-20b. Mechanistically, CDC37L1 binds to the heat shock protein HSP90 to activate the peptidyl-prolyl cis-trans isomerase A (PPIA) that accelerates protein folding; higher mRNA levels associated with poorer OS and DFS in sorafenib-treated patients [
51]. Another study employed a CRISPR-based screening method in vivo by using a sorafenib-treated xenograft model to improve the translational value of preclinical data with respect to in vitro tools. The Authors identified miR-3689a-3p as the most overexpressed miRNA in sorafenib-sensitive tumors and reported the targeting of the copper chaperone for superoxide dismutase (CCS) which, by reducing SOD1 ability to scavenge mitochondrial ROS, increased cellular oxidative stress that eventually mediated the antitumor effect of sorafenib. Orthotopic mouse models showed that miR-3689a-3p downregulation decreased sorafenib efficacy. Since lower miR-3689a-3p levels were detected in tumor specimens from HCC patient cohorts, this study paves the way towards a combined miRNA mimics and sorafenib strategy to boost sorafenib anticancer efficacy [
52]. Notably, this high throughput screening procedure is particularly suitable for the discovery of driver genes and therapeutic targets that modulate drug efficacy. An example is represented by the metabolic gene phosphoglycerate dehydrogenase (PHGDH), regulating the serine synthesis pathway, whose specific inhibition by NCT-503 acted synergistically with sorafenib to abolish in vivo tumorigenesis [
53]. Similarly, the downregulation of Kelch-like ECH-associated protein 1 (KEAP1) in response to sorafenib administration increased the activity of Nrf2, a key transcription factor controlling antioxidant responses, which contributed to enhance drug resistance to sorafenib, lenvatinib, and regorafenib in HCC [
54].
Metabolic reprogramming from oxidative phosphorylation to aerobic glycolysis, also known as ‘Warburg effect’, is a core hallmark of cancer cells [
55] influencing the response to sorafenib. Even though ATP production during aerobic glycolysis is much lower, the Warburg effect confers advantages to cancer cells growth by providing the carbon sources required for rapid cell proliferation and, in the meantime, by minimizing the production of toxic ROS [
56]. Several HCC-specific miRNAs are involved in this glycolytic shift, as in the case of miR-3662, which is downregulated in liver tumors. Its reinforced expression in HCC cell lines associated with a decrease in glucose and oxygen consumption, ATP and lactate production, and in vivo tumorigenesis. Interestingly, both HIF-1A and esokinase 2 (HK2) are direct targets of this miRNA and their overexpression mitigates the above-mentioned effects confirming their key activity in mediating miR-3662 biologic processes [
57,
58]. We recently reported that the oncomiR-494 can rewire tumor metabolism of HCC cells by targeting the catalytic subunit of Glucose-6 phosphatase (G6pc) which is a multi-subunit complex catalyzing the de-phosphorylation of G6P to free glucose playing a central role in glucose homeostasis. A negative correlation was displayed between miR-494 and G6pc in HCC patient cohorts where lower G6pc levels associated with high tumor grade, microvascular invasion (MVI) and larger tumor size. We demonstrated that miR-494/G6pc axis contributes to metabolic plasticity of cancer cells favoring the accumulation of glycogen and lipid droplets that are exploited in case of critical metabolic conditions (e.g., glucose deprivation) giving an advantage to uncontrolled proliferation of malignant cells. We also showed that miR-494/G6pc axis promotes sorafenib resistance and proposed combined antagomiR-based treatments with sorafenib or 2-deoxyglucose (2-DG) for HCC patients who may develop sorafenib resistance and that are ineligible for immunotherapy [
59]. An interesting study by Zhang et al. [
60] demonstrated the pivotal role of miR-30a-5p/CLCF1 axis in modulating the metabolic shift toward aerobic glycolysis of SR HepG2 cells and xenograft tumors. Specifically, they found a time-dependent decrease of miR-30a-5p in sorafenib treated cells and demonstrated by functional analysis the direct targeting of the pro-inflammatory cytokine CLCF1 that activates the downstream PI3K/AKT pathway controlling proliferation and metabolic reprogramming of cancer cells. Indeed, the treatment with the AKT inhibitor MK-2206 reverted the glycolytic phenotype of SR HepG2 cells decreasing ATP and lactate production as well as mRNA expression of metabolic genes GLUT3, HK2, and PDK1. Strikingly, the Authors proved the therapeutic efficacy of a lipid formulation containing a chemically modified oligonucleotide (2′-O-methyl-modified miRNA conjugated with cholesterol) mimicking miR-30a-5p, which was injected into the tail vein of immunocompromised mice (once a week for five weeks). This miRNA formulation effectively inhibited the tumor growth of SR HepG2 cells, proving the feasibility and safety of miRNA delivery in vivo and its efficacy against sorafenib resistant tumors. An inverse correlation between miR-30a-5p and CLCF1 was found in HCC patients confirming the importance of this signaling axis in human tumors and suggesting the combination of agomiR-30a-5p and sorafenib as a promising strategy to improve TKIs efficacy and overcome acquired resistance.
In a treatment perspective, small extracellular vesicles (EV), which are loaded with miRNAs, proteins, and mRNAs protecting them from degradation, represent promising drug delivery vehicles and ideal miRNAs carriers to cancer cells [
61]. Mesenchymal stem cells are a precious source of EVs retaining the characteristics of their parental cells and showing low immunogenicity and tumor-delivery properties [
62]. An elegant study by Sun et al. described the engineering of EVs with miR-654-5p by in vitro electroporation (m654-sEV) and reported their effectiveness in sorafenib sensitization in preclinical models derived from SR resistant HCC cells through the direct targeting of the ferroptosis inhibitor HSPB1 [
63]. In vivo findings proved that the combination of sorafenib and m654-sEV strongly suppressed tumor growth in comparison to sorafenib treatment alone by modulating ferroptosis-associated markers. In particular, the combined treatment effectively inhibited HSPB1 expression, increased levels of transferrin receptor (TFRC), COX2, Fe2+, and ROS, together with a decrease in glutathione (GSH) levels, suggesting this strategy as a reliable one to overcome sorafenib resistance in HCC. Another miRNA involved in sorafenib resistance via impairment of iron-associated programmed cell death, ferroptosis, is miR-23a-3p which is overexpressed in sorafenib-resistant patients and associated with tumor recurrence. A sorafenib resistant xenograft model obtained by inoculation of MHCC97L cells showed that miR-23a-3p is overexpressed in tumors acquiring resistance after long drug exposure. Tumor-derived resistant cell lines displayed a transcriptional activation of pri-miR-23a mediated by ETS1 transcription factor. A consistent reduction in cell growth was obtained in an orthotopic model when miR-23a-3p knockout HCC cells where injected in the presence of sorafenib treatment. A functional analysis assessed the targeting of Acyl-CoA synthetase long-chain family member 4 (ACSL4), a necessary enzyme for catalyzing lipid peroxidation during ferroptosis, suggesting the silencing of miR-23a-3p as a promising option in sorafenib resistant HCC patients [
64].
The reactivation of oncogenic pathways is a common mechanism of drug resistance to TKIs in HCC [
65] and PI3K/AKT alterations might predict sorafenib resistance [
10]. We described the dual role of miR-30e-3p on tumorigenesis and sorafenib resistance based on TP53 status [
29]. We showed that miR-30e-3p behaves as a TS miRNA in p53 wild type cells establishing a feedforward loop with TP53/MDM2 axis while it behaves as an oncogene in p53 mutated backgrounds targeting PTEN/AKT pathway and driving drug resistance. In the DEN-HCC rat model treated with sorafenib, which highly mirrors the human disease [
66], a lower miR-30e-3p expression was detected in non-responder tumors displaying a negative correlation between miR-30e-3p and tumor size and a positive correlation with apoptotic markers, demonstrating the involvement of this miRNA in sorafenib sensitization. Another study reported the downregulation of miR-124-3p.1 in liver tumors and described its role in sorafenib response by targeting SIRT1 and AKT2 preventing nuclear translocation of FOXO3a transcription factor. Treatment combination between miR-124-3p.1 mimics and sorafenib improved its antitumor effect in a nude mouse model [
67]. Several evidence reported the upregulation of the IGF/FGF pathways during acquired resistance to sorafenib [
68,
69]. Lin and colleagues investigated the mechanisms that lead to miRNA deregulation in SR cells. They identified the downregulation of exportin 5 (XPO5) via DNA promoter methylation to be responsible for impaired miR-378a maturation driving IGF1R signaling activation [
70]. The anti-cancer strategy pursued by these Authors took advantage of GW3965, an agonist molecule of the transcription factor LXRα which mediates miR-378a transcription. Sorafenib plus GW3965 therapy demonstrated a consistent inhibition of tumor growth compared with sorafenib alone in both orthotopic and patient-derived xenograft (PDX) mouse models demonstrating the regulation of miRNA biogenesis as a promising option to improve sorafenib effectiveness in HCC. FGFR4 and EGFR oncogenes are upregulated in SR resistant cell lines and are direct targets of miR-486-3p which is downregulated in liver tumors and correlates with poor survival [
71]. Intratumor injection of lentiviral particles carrying miR-486-3p in sorafenib resistant SK-Hep1-derived orthotopic mice synergistically improved sorafenib efficacy. Similarly, circular RNA named circRNA-SORE resulted upregulated in SR HCC cells, xenograft and PDX models due to increased N6-methyladenosine (m6A) levels that positively influenced its mRNA stability. Lower circRNA-SORE associated with better OS and recurrence-free survival in sorafenib-treated patients. It acted as a ceRNA by sequestering miR-103a-2-5p and miR-660-3p, promoting Wnt/β-catenin pathway activation that triggers and maintains a drug resistant phenotype. Orthotopic models with sorafenib resistant SK-Hep1 cells silenced for circRNA-SORE displayed a higher sensitization to sorafenib treatment. In agreement, intratumor injection of short hairpin (sh-circRNA-SORE) lentiviral particles in sorafenib resistant HCCLM3-derived xenograft mice potentiated the antitumor effect of sorafenib, suggesting the clinical potential of ncRNAs-based combined strategies [
72]. The only concern relative to the last two studies regards the use of the SK-Hep1 cell line for the establishment of orthotopic animal models. Indeed, SK-Hep1 cells originate from liver endothelial cells and not from parenchymal tumor hepatocytes. The use of inappropriate animal models may be one of the causes affecting preclinical data translation into the clinical practice, therefore particular attention should be paid when choosing preclinical tools. Circular RNA cDCBLD2 is upregulated in SR cell lines where it sponged miR-345-5p increasing type IIA topoisomerase expression (TOP2A) that reduced the sorafenib-mediated apoptotic effect. Higher TOP2A expression associated with recurrence, and metastasis of HCC patients treated with sorafenib and with worse OS and recurrence-free survival. Local injection of cholesterol-conjugated small interfering RNA molecules (si-cDCBLD2) in a sorafenib-resistant PDX model increased drug sensitivity, supporting the clinical potential of cDCBLD2 silencing to enhance sorafenib efficacy in resistant patients [
73].
Regarding the role played by autophagy on sorafenib resistance, Li and coworkers reported the nuclear activation of the lncRNA SNHG1 by miR-21 in SR HCC cells and described the activation of the AKT pathway via SLC3A2 upregulation [
74]. Interestingly, in vitro inhibition of this lncRNA by an anti-SNHG1 siRNA strategy induced sorafenib sensitization through the activation of autophagy and apoptotic cascade; moreover, it showed tumor inhibition in vivo exerting a synergistic effect with sorafenib co-administration. On the contrary, miR-541 sensitized HCC cells to sorafenib treatment by inhibiting the expression of two autophagy-related genes, ATG2A and RAB1B, highlighting the opposite role attributed to autophagy on drug sensitization [
75]. MiR-541 is downregulated in HCC and its low expression correlates with shorter OS and high recurrence rate and predicts sorafenib resistance. Notably, intratumor injection of Ad-miR-541 potentiated the effects of sorafenib in xenograft mice resulting in maximal tumor growth inhibition [
76].
Considering the central activity of liver cytochrome P450 family in drug metabolism, He et al. investigated the effect of CYP3A4 on sorafenib metabolism and clearance and displayed that a higher expression associates with poor survival in sorafenib-treated patients [
77]. The Authors demonstrated CYP3A4 targeting by miR-4277 and reported an addictive antitumor effect of miRNA mimics plus sorafenib in immunocompromised mice. To this regard, the study by Li and colleagues dissected the mechanisms downstream the decreased expression of miR-138-1-3p in HCC and found the serine/threonine kinase PAK5 among its targets. PAK5 upregulation triggered β-catenin phosphorylation causing its nuclear translocation which, in turn, activated the transcription of the multidrug resistant transporter ABCB1 responsible for sorafenib efflux and decreased effect. Notably, combined treatments with lentiviral vectors for miR-138-1-3p or PAK5 shRNA together with sorafenib had an enhanced anticancer effect with respect to sorafenib monotherapy in SR HepG2-derived xenograft mice [
78].
In conclusion, ncRNAs regulate sorafenib resistance through a variety of molecular mechanisms (
Figure 1). Preclinical studies demonstrated that combined strategies designed to restore the deregulated expression of ncRNAs enhance sorafenib sensitization in HCC, opening the path towards the design of focused clinical trials to improve treatment efficacy and patient survival.
3.2. Non-coding RNAs and lenvatinib combination improves the therapeutic response
Lenvatinib is an oral TKI targeting VEGFR, FGFR, PDGFRa, RET, KIT [
79]. A randomized phase III clinical trial demonstrated that lenvatinib is non-inferior to sorafenib, showing an OS of 13.6 months; therefore, it granted approval as first line treatment in 2018 [
4]. However, only a low percentage of advanced HCC patients benefit from lenvatinib, with the great majority being non-responder or developing drug resistance before or during treatment [
80]. For this reason, the knowledge of the underlying molecular mechanisms and the discovery of new target genes for combination strategies are urgent clinical needs aimed at improving lenvatinib efficacy.
Wei et al. showed that miR-3154 influence lenvatinib response, being upregulated in lenvatinib-resistant (LR) HCC cells. To validate this hypothesis, miR-3154 was silenced in HCC cells treated with lenvatinib, resulting in reduced cancer stem cell markers, colony formation and increased apoptosis. These effects were confirmed in PDX mouse models, where tumor volume was reduced upon lenvatinib treatment in low miR-3154 tumors only. Mechanistically, miR-3154 target the transcription factor hepatocyte nuclear factor 4 alpha (HNF4α) which is indispensable for hepatocyte differentiation and critical for maintaining liver health, preventing its nuclear translocation. Moreover, in a cohort of HCCs receiving lenvatinib after surgical resection, patients with low miR-3154 levels had a better survival compared to those bearing high levels. Considering that low miRNA levels correlate with a better response, the preliminary evaluation of miR-3154 in HCC tissue could help identify in advance patients who may benefit from lenvatinib before treatment start. On the other hand, if confirmed by other studies, miR-3154 could represent a therapeutic target improving lenvatinib sensitivity in HCC [
81]. MiR-183-5p.1 promotes the expansion of liver tumor-initiating cells (T-ICs) by regulating the expression of MUC15, a membrane-associated mucin whose downregulation was previously associated with advanced stages, poorly differentiated and metastatic liver cancers [
82]. Han et al. showed that downregulation of MUC15 elevated the expression of T-IC associated markers, promoting malignant transformation of hepatocytes and spheroid formation in vitro. Consistently, downregulation or deletion of MUC15 in murine models dramatically increased tumor number, size, and liver-to-body weight ratio. These effects were mediated by increased levels of c-MET, PI3K and p-AKT, revealing the existence of a miR-183-5p.1/MUC15/c-MET/PI3K/AKT/SOX2 regulatory circuit in liver T-ICs. In line with these results, miR-183-5p was upregulated in HCCs compared with normal tissues. On the contrary, HCC patients with high MUC15 expression displayed a prolonged survival following lenvatinib treatment, suggesting its evaluation as a predictor of lenvatinib response. In agreement, patient-derived organoid (PDO) and PDX models expressing low MUC15 levels were resistant to lenvatinib treatment. Given these evidence, the administration of anti-miR-183-5p.1 could be a possible strategy to increase MUC15 levels in HCC patients treated with lenvatinib [
83]. Another miRNA involved in the regulation of oncogenic pathways is miR-128-3 that mediates lenvatinib resistance in HCC cells by downregulating c-Met [
84]. In LR HCC cells, miR-128-3p mimics strengthened the anti-proliferative effects of lenvatinib by directly targeting c-Met resulting in the downregulation of the ERK/cyclin D1 pathway, which is involved in cell cycle progression. In addition, miR-128-3p mimics enhanced lenvatinib-induced apoptosis in LR-HCC cells through the downregulation of p-Akt and p-GSK-3β, and the increase of caspase-9 and -3 cleavage. In xenograft mice injected with LR HCC cells, both lenvatinib treatment and miR-128-3p mimics resulted in significantly smaller tumors compared to controls. Notably, the combination therapy led to even smaller tumors than each monotherapy, showing higher apoptosis and lower proliferation indexes together with reduced p-Akt and p-GSK-3β expression. These evidence suggest that the combinatorial therapy of lenvatinib plus miR-128-3p mimics could be explored in clinical trials to further increase the efficacy of lenvatinib and possibly overcome the development of resistance.
Liu et al. revealed that low basal circKCNN2 levels associate with worse prognosis and tumor recurrence in HCC patients but, on the other side, predispose to a stronger anti-tumor effect of lenvatinib via miR-520c-3p/MBD2 axis [
85]. Mechanistically, by sponging miR-520c-3p, circKCNN2 avoids its binding to methyl-DNA-binding domain protein 2 (MBD2), resulting in reduced proliferation, migration, colony formation and cell cycle progression in HCC cells, and lower tumor burden in vivo. Moreover, cells and PDOs with lower intrinsic circKCNN2 levels were more sensitive to lenvatinib treatment but have a higher risk of tumor recurrence. On the contrary, ectopic expression of circKCNN2 together with lenvatinib treatment have synergistic effects, possibly because they both downregulate the FGF19/FGFR4/FRS2 pathway. Indeed, circKCNN2 represses FGFR4 through miR-520c-3p/MBD2 axis. In turn, intrinsic high levels of circKCNN2 may reduce the effectiveness of lenvatinib because the FGF19/FGFR4/FRS2 pathway is already inhibited. Conclusively, this work revealed that circKCNN2 may be a promising predictive biomarker of HCC recurrence and treatment sensitivity, as well as a therapeutic agent in low-expressing patients in combination with lenvatinib, even though caution should be paid due to its dual role on drug sensitivity and tumor recurrence.
MT1JP is a lncRNA acting as a ceRNA for miR-24-3p. Yu et al. found the upregulation of MT1JP in LR HCC cells and showed that lenvatinib itself promotes MT1JP expression in vitro. Conversely, viability and apoptosis assays showed that the overexpression of miR-24-3p sensitizes HCC cells to lenvatinib. To better understand the molecular mechanisms governing MT1JP/miR-24-3p-mediated lenvatinib response, the authors demonstrated that the antiapoptotic factor BCL2L2 is a miR-24-3p target gene, and its expression confers a survival advantage to lenvatinib-treated cells. In PDXs treated with lenvatinib, responder tumors had low MT1JP and high miR-24-3p levels, together with increased apoptotic markers, compared to non-responders. Moreover, injection of MT1JP-overexpressing SMMC-7721 cells gave rise to bigger tumors in lenvatinib-treated xenograft mice. These data suggested that MTJ1P silencing or miR-24-3p mimics could be used as co-treatments to increase lenvatinib efficacy in HCC [
86]. Another lncRNA upregulated in LR HCC cells, PDOs and patients is LINC01607. Notably, a significant reduction of ROS production was found in an orthotopic HCC model following LINC01607 overexpression being responsible for in vivo lenvatinib resistance. Mitophagy was activated following lenvatinib treatment, suggesting its contribution to the enhanced antioxidant capacity of LR HCC cells helping them to maintain low oxidative stress levels. To explain molecular mechanisms underneath LINC01607 deregulation, it emerged that it acts as a ceRNA for miR-892b increasing p62-associated mitophagy. P62 also regulates Nrf2 expression, which in turn protects cancer cells from oxidative stress regulating the expression of several antioxidant genes. Finally, a xenograft model with LR Hep3B cells silenced for LINC01607 demonstrated its synergistic effect with lenvatinib treatment and the same was confirmed in the PDO model. Taken together, these results indicated that LINC01607 promotes antioxidant capacity of LR HCC cells through miR-892b/p62/Nrf2 axis [
87], suggesting non-coding RNAs as promising therapeutic targets to overcome lenvatinib resistance in HCC.
Given the central role of miRNAs in regulating HCC progression and response to treatment, they were also explored as therapies on their own. This is the case of miR-22, whose reduced expression was linked to poor survival outcome in patients with HCC. Hu et al. proved that miR-22 gene therapy is an effective treatment in two orthotopic HCC mouse models, ensuring prolonged survival compared to lenvatinib, without causing detectable toxicity [
88]. The anti-HCC effects of miR-22 were mediated by its immunomodulatory functions on T cells. Indeed, miR-22 silenced HIF-1A and increased retinoic acid (RA) signaling in both hepatocytes and T cells, therefore repressing IL-17 pro-inflammatory signaling and inhibiting Th17 and Treg cells expansion, while enhancing cytotoxic CD8+ T cells recruitment, activation, and survival. Additionally, miR-22 treatment improved metabolism, inhibited inflammation, and reduced hypoxia signaling. These data suggested miR-22 gene therapy as a novel effective option for HCC treatment that may also empower the effect of immunotherapy by favoring a cytotoxic immune response against HCC. There is evidence of using lenvatinib as second line therapy for HCC patients undergoing sorafenib resistance. In this context, Shi et al. underlined the importance of considering lenvatinib influence on miRNAs expression profile in SR Huh-7 cells, thus identifying possible targets influencing HCC cells sensitivity to TKIs. For instance, lenvatinib treatment reduced the expression of two HCC-associated miRNAs (miR-130b and miR-106b) whose high levels associated with reduced OS in HCC patients. For this reason, studying this aspect and its molecular implications in depth could be of great significance to improve HCC management in sorafenib-resistant patients [
89].
In summary, despite the low number of studies reporting mechanisms underneath lenvatinib resistance, here we showed that a variety of ncRNAs modulate lenvatinib response in HCC preclinical models (
Figure 2) deserving attention as promising candidates for combined strategies.