2023 Impact Factor
Small interfering RNA (siRNA) consists of a double-stranded RNA, including a passenger (sense) strand and a guide (antisense) strand, each approximately 19-23 base pairs in length with 3’ dinucleotide overhangs (Friedrich and Aigner, 2022). Typically, synthetic exogenous siRNAs are delivered to cells via liposomes, electroporation, or conjugation with fatty acids or other moieties. Once inside the cell’s endosome, siRNA must escape into the cytoplasm and integrate into the RNA-induced silencing complex (RISC). The siRNA is activated by removing the passenger strand, while the guide strand directs the RISC to bind to the target mRNA in a sequence-specific manner and degrade it (Hu
In addition to 100% complementarity binding-mediated degradation of target mRNAs, siRNAs can also bind to target and nontarget mRNAs via partial complementarity, resulting in translational repression and off-target effects. These effects can be caused by both sense and antisense strands and necessitate mechanisms to mitigate the seed-mediated off-target effects of siRNAs. Several strategies can reduce these off-target effects, including the design of asymmetric siRNAs (asiRNAs), modification of ribose sugars, and pooling multiple siRNAs targeting the same mRNA (Biscans
The use of asiRNAs offers many benefits, including the reduction of passenger strand seed-mediated off-target effects, the possible enhancement of passive cellular uptake, and the recognition of asiRNA overhangs by the RISC (Biscans
A major hurdle in the clinical development of siRNA-based drugs is the lack of a proper delivery system. Recently, various siRNA delivery systems have been used in clinical applications, especially for targeting liver diseases. Although the delivery system for liver diseases has been resolved, the delivery of siRNA to extrahepatic tissues remains a challenge. Previous reports have shown that docosanoic acid (DCA)-conjugated asiRNAs are superior in terms of safety and knockdown efficacy (Biscans
Breast cancer is a highly heterogeneous disease that is broadly classified into four major subtypes: luminal A, luminal B, Her2 enriched, and TNBC (Orrantia-Borunda
In this study, we aimed to target the
siRNAs were designed using the freely available software siDirect v2.0 (Naito
The triple-negative breast cancer cell lines MDA-MB-157, MDA-MB-231, MDA-MB-436 and Hs-578T were obtained from the Korean Cell Line Bank and used immediately. Prior to use, all cell lines were tested for mycoplasma contamination using the CycleavePCR™ Mycoplasma Detection Kit (#CY232, TaKaRa Bio Inc., Shiga, Japan). MDA-MB-157, MDA-MB-231, and MDA-MB-436 cells were cultured in RPMI 1640 medium (#LM 011-01, WELGENE, Gyeongsan, Korea), while Hs-578T cells were cultured in DMEM supplemented with high-glucose Dulbecco’s modified Eagle’s medium (#001-05, WELGENE). Both media were supplemented with 10% fetal bovine serum (FBS), 100 units/mL penicillin, and 100 μg/mL streptomycin. All cultures were maintained at 37°C in a humidified atmosphere of 95% air and 5% CO2.
A total of 1.5-2×105 cells were seeded in each well of a six-well plate and cultured overnight at 37°C and 5% CO2. The following day, upon reaching 60-80% confluency, the cells were washed with phosphate-buffered saline (PBS), and the medium was replaced. The transfection mixture was prepared by combining 200 μL of Opti-MEM™ I Reduced Serum Medium (#319850620, Thermo Fisher Scientific, Carlsbad, CA, USA), 5-7 μL of Lipofectamine™ 3000 (#L3000015, Invitrogen, Carlsbad, CA, USA) or DharmaFECT 1 (#T-2001-03, Horizon Discovery, Cambridge, UK), and 2 μL of 20 μM siRNA solution in a 1.5 mL Eppendorf tube. The mixtures were incubated for 15 min at room temperature with Lipofectamine™ 3000 and for 20 min with DharmaFECT 1 (#T-2001-03, Horizon Discovery). Following incubation, 207-209 μL of the transfection mixture was added to the corresponding wells of the six-well plates. The plates were then incubated for 48-72 h at 37°C and 5% CO2.
Cells were cultured in six-well plates and treated with specific siRNAs. The plates were incubated at 37°C and 5% CO2 for 48-72 h. Following the incubation period, the culture medium in each well was replaced with 1800 μL of fresh medium. Subsequently, 200 μL of EZ-Cytox cell viability reagent (#EZ-3000, Doogenbio Co., Ltd., Suwon, Korea) was added to each well. The plates were then incubated for an additional 3 h at 37°C. After incubation, 100 μL of the medium from each well was carefully transferred to the corresponding wells of a 96-well plate, and the assay was performed at least in duplicate. The absorbance was measured at a wavelength of 450 nm using a SpectraMAX M5 Multi-Reader (Molecular Devices, San Jose, CA, USA). The data were analyzed in Excel, with all treatment groups normalized to either nontreated or mock or negative control (scramble) siRNA.
Cell lysis was performed using RIPA lysis buffer (#89901, Thermo Fisher Scientific, San Francisco, CA, USA) supplemented with Halt™ Protease and Phosphatase Inhibitor Cocktail (100×) (#78440, Thermo Fisher Scientific). Protein concentrations were determined by Pierce™ BCA Protein Assay Kits (#23225, Thermo Fisher Scientific). The cell lysates were resuspended in 5x SDS‒PAGE loading buffer (#SF2002-110-00, Biosesang, Yongin, Korea) and distilled water and then heated at 95°C for 5 min.
The protein lysates were resolved by 10-12% SDS‒PAGE. Each gel was loaded with 20-35 µg of protein and electrophoresed for 20 min at 80 volts through the stacking gel, followed by approximately 90 min at 100 volts through the separating gel. Proteins were transferred to a 0.2 µm polyvinylidene difluoride (PVDF) membrane at 100 volts for 90 min using 5.8 g of Trizma® base transfer buffer (#T1503-1KG, Sigma‒Aldrich, St. Louis, MO, USA), 29 g of glycine (#01194-00, Samchun Chemicals Co., Ltd., Seoul, Korea), 400 mL of 99.5% methyl alcohol (#M0585 Samchun Chemicals. Co., Ltd.), and distilled H2O to make 2 liters. Blocking was performed with 5% skim milk (#232100, BD Biosciences, Sparks, MD, USA) in 1× TBST for 30 min on a shaker at room temperature. Primary and secondary antibodies were diluted within the recommended ranges in 1% skim milk with 1× TBST. After blocking, the membrane was washed once, and primary antibodies were applied and incubated overnight at 4°C on a shaker. The membranes were then washed three times for 10 min each for a total of 30 min while shaking. The membranes were incubated with secondary antibodies for 2 h at room temperature on a shaker. Subsequent washes involved three cycles of 10 min each in 1× TBST, totaling approximately 30 min. Image documentation was performed by an LAS4000 instrument (ImageQuant, GE Healthcare, Hertfordshire, UK) using chemoluminescence, with the help of detection reagent A (#RPN2232S1: luminol enhancer solution from GE Healthcare) and reagent B (#RPN2232S2: peroxide solution from GE Healthcare), which were mixed at a ratio of 1:1.
The following primary antibodies were used: recombinant anti-c-MYC antibody [Y69] (#ab32072, Abcam, Cambridge, MA, USA), anti-beta actin antibody (#ab8227, Abcam); anti-GAPDH antibody [EPR16891] as a loading control (#ab181602, Abcam), anti-RAD51 antibody ([EPR4030(3)] (#ab133534, Abcam), PARP1 (46D11) rabbit mAb (#9532, Cell Signaling Technology, Danvers, MA, USA) and RRM2 (E7Y9J) XP® rabbit mAb (#65939, Cell Signaling Technology). The secondary antibody used was goat anti-rabbit IgG H&L (HRP) (#ab205718, Abcam).
Total RNA was isolated from cell lines and tissues using Hybrid-RTM (#305-101, GeneAll Biotechnology, Seoul, Korea) with RiboExTM lysis. The concentration of total RNA was determined by measuring the absorbance at 260 nm (A260) using a NanoDrop device (Thermo Fisher Scientific). The purity of the RNA was assessed by the A260/A280 ratio. For cDNA synthesis, 1 μg of total RNA was reverse transcribed using a RevertAid First Strand cDNA Synthesis Kit (#K1622, Thermo Fisher Scientific). The master mix for reverse transcription included 1 μL of a mixture of Oligo (dT)18 and random hexamer primers. This mixture (2 μL) was added to each RNA sample tube. The RNA samples were denatured at 65°C for 5 min and then allowed to anneal at 40°C for 5 min. The second component of the master mix was prepared by combining 4 μL of 5× reaction buffer, 1 μL of RiboLock RNase inhibitor, 2 μL of 10 mM dNTP mix, and 1 μL of RevertAid M-MuLV RT (200 U/μL), totaling 8 μL per sample. The reaction conditions were set on a TaKaRa Thermal Cycler Dice Touch (TaKaRa Bio Inc., Shiga, Japan) at 25°C for 5 min, 42°C for 1 h, and 70°C for 5 min, followed by a hold at 4°C.
qPCR was conducted using TOPrealTM SYBR Green qPCR 2X PreMIX with low ROX (#RT500M, Enzynomics, Daejeon, Korea) on a 7500 Real-Time PCR system (Applied Biosystems, Carlsbad, CA, USA; Thermo Fisher Scientific). Primers were designed using the PrimerQuest™ Tool available on the Integrated DNA Technologies (IDT, Coralville, IA, USA) website, as detailed in Table 1. The qPCR protocol was as follows: initial denaturation at 95°C for 12 min, followed by 45 cycles of denaturation at 95°C for 10 s, annealing at 60°C for 15 s, and elongation at 72°C for 15 s. HPRT1 served as the endogenous control for normalization. Ct values were analyzed using the delta-delta Ct (2^−ΔΔCq) method (Livak and Schmittgen, 2001).
Table 1 Primers used for mRNA quantification by qPCR
Gene name | Primers | (5'→3') | |
---|---|---|---|
1 | PARP1 | Forward primer | GGA TCA GGG TGT AAA AGC GAT |
Reverse primer | CGC ATA CTC CAT CCT CAG TG | ||
2 | MYC | Forward primer | TCT TCC TCA TCT TCT TGT TCC TC |
Reverse primer | TCC TCG GAT TCT CTG CTC TC | ||
3 | RAD51 | Forward primer | ACA TTA TCC AGG ACA TCA CTG C |
Reverse primer | GCC ATG TAC ATT GAC ACT GAG | ||
4 | HPRT1 | Forward primer | GCG ATG TCA ATA GGA CTC CAG |
Reverse primer | TTG TTG TAG GAT ATG CCC TTG A | ||
5 | RRM2 | Forward primer | TCTTGCATTGTGAGGTACAGG |
Reverse primer | TCTGAGCTGGCAGAAGTTAATC |
To assess the serum stability of the siRNAs, 6 μL of 20 μM siRNA (~1.5 μg) was incubated in 8 μL of 100% fetal bovine serum (FBS) as described previously (Jung
All animal procedures were approved by the Institutional Animal Care and Use Committee (IACUC) of Seoul National University (SNU-210721-1). In this study, 6-week-old female BALB/c-nude mice were housed in ventilated cages with ad libitum access to food and water, a 12 h light/dark cycle, and controlled temperature and humidity in an animal facility. Tumor xenografts were generated by subcutaneously (SC) injecting 100 μL of 6.5×106 MDA-MB-231 cells prepared in 50 μL of serum-free RPMI 1640 (#LM 011-01, WELGENE) mixed with 50 μL of BD Matrigel matrix at a high concentration (#354248, BD Biosciences, Plymouth, UK) in the right flank of 14 mice. Once the tumor diameter reached greater than 300 mm3, the mice were administered a single dose of 20 mg/kg asiRNA-VP-DCA, scramble asiRNA-VP-DCA or the same volume of PBS via the SC route. Mice were euthanized on days 2, 7, 14, and 28 post-injection, and tumor tissues were collected in a cryovial and preserved in RNAlater™ Solution (RNA stabilization solution, #AM7020, Invitrogen, Vilnius, Lithuania) for RT‒qPCR analysis or snap-frozen in liquid nitrogen for western blot analysis. Both western blotting and qPCR experiments were conducted as described for the
In addition, 20 BALB/c nude mice were injected SC in the right flank of MDA-MB-231 cells to investigate the long-term knockdown effects of repeated doses. Once the tumor volume exceeded 150 mm3, the mice were randomly assigned to one of 4 groups: PBS (SC), asiRNA-VP-DCA (SC), scramble-asiRNA-VP-DCA (SC), and asiRNA-VP-DCA (IT). Mice received injections of 100 μL of either PBS (SC) or 100 μL of 15 mg/kg asiRNA-VP-DCA (SC) targeting
Data analysis was performed using GraphPad Prism version 8.0.2 software (Dotmatics, Boston, MA, USA). One-way and two-way ANOVA with Tukey’s and Sidak’s multiple comparisons tests were used to determine the significance level between treatment groups. The IC50 curves were fitted using log (inhibitor) versus normalized response with variable slope. In all cases, the significance level was set at the 95% confidence interval with *
Prototype siRNA design (19+2/19+2): A total of 12 siRNAs targeting the
After the initial screening, to enhance serum stability, potency, safety, and durability, one siRNA candidate, referred to as siRNA8, was selected for further modifications. These modifications and conjugations include 2-Ome + 2’-F (Fig. 1B), 2’-Ome + 2’-F + PS (Fig. 1C), and 2’-Ome + 2’-F + PS + ChoL conjugation at the 3’ end of the passenger strand using a noncleavable linker (triethylene glycol) (Fig. 1D).
Asymmetric siRNA design: To mitigate seed-mediated off-target effects and enhance specificity, we systematically designed a series of asiRNAs by progressively shortening the passenger strand. Starting with a standard 19+2 nucleotide configuration on the guide strand, we reduced the length of the passenger strand by one nucleotide, resulting in configurations such as 19+2/18+2 (Fig. 1E), 19+2/17+2 (Fig. 1F), 19+2/16+2 (Fig. 1G), 19+2/15+2 (Fig. 1H), 19+2/14+2 (Fig. 1I), and 19+2/13+2 (Fig. 1J).
Additionally, to explore the impact of eliminating the 2’-overhang on the passenger strand, we designed configurations such as 19+2/16+0 (Fig. 1K), 19+2/15+0 (Fig. 1L), and 19+2/15+0, with further modifications by removing one nucleotide from the 5’ end and five nucleotides from the 3’ end of the passenger strand, as depicted in Fig. 1M.
Furthermore, because shorter asiRNAs may further minimize off-target effects, we synthesized an asiRNA with an overall length of 20 nucleotides. These included 18+2/15+2 (Fig. 1N) and 18+2/15+0 (Fig. 1O).
Modified and conjugated asymmetric siRNA design: Following the initial screening of various asiRNAs for their MYC knockdown efficacy, the 19+2/16+2 design was selected for further modifications, including 2’-Ome + 2’-F + PS + VP (Fig. 1P). This modified asiRNA was then validated for its
While the extensive modification of siRNA is typically advantageous for enhancing nuclease resistance and ensuring durable efficacy, heavy modifications at the 5’ end (e.g., a combination of 2’-Ome or 2’-F with PS) and a high proportion of 2’-F modifications have occasionally led to interference and long-term toxicity, respectively.
Given these challenges and considering that 2’-Ome modifications are known to be more stable than 2’-F modifications, we developed additional modification strategies. These included reducing the modifications at the 5’ end of the guide strand (specifically omitting PS modifications) and decreasing the overall number of 2’-F modifications. These adjustments aim to reduce off-target effects and long-term toxicity associated with fluorine modifications while enhancing the nuclease stability of 2’-Ome over 2’-F modifications (Fig. 1R, 1S).
Initial screenings were conducted using MDA-MB-157, MDA-MB-231, and MDA-MB-436 cells, each treated with 20 nM siRNA. Twelve siRNAs were evaluated for their ability to knockdown mRNA and protein expression (Fig. 2A-2C). siRNA7 through siRNA12 achieved greater than 50% mRNA knockdown efficacy in at least one of these cell lines (Fig. 2B) and were selected for further analysis. In contrast, siRNA1 through siRNA6 were excluded from subsequent tests due to insufficient efficacy.
Further screening via western blotting revealed that siRNA8 and siRNA10 had superior knockdown efficacy across all three cell lines (Fig. 2C), indicating that western blotting provided more specific results than mRNA quantification at this screening stage. Various factors, including the dose of siRNA, volume of transfection reagent, assay timing post-transfection, and cell count, significantly influenced the knockdown efficiency (Fig. 2D-2H). Specifically, increasing the siRNA dose (Fig. 2D) and the volume of the transfection reagent (Fig. 2E) significantly improved the mRNA knockdown efficacy. Conversely, a greater cell number reduced the efficacy (Fig. 2F), while increasing the volume of DharmaFECT 1 enhanced it (Fig. 2F). Additionally, no significant differences in knockdown efficacy were observed between the DharmaFECT 1 and Lipofectamine™ 3000 transfection reagents in Hs-578T cells (Supplementary Fig. 1). Notably, the knockdown efficacy of siRNAs at both the mRNA and protein levels improved over time; for example, assays conducted 72 h post-treatment showed significantly greater mRNA knockdown in MDA-MB-157 cells than at 24 h (Fig. 2G, 2H).
Next, we evaluated the silencing efficacy of modified and/or conjugated siRNA8 derivatives, siRNA8-mf (2’-Ome + 2’-F), siRNA8-mf-PS (2’-Ome + 2’-F + PS), and siRNA8-mf-PS-ChoL (2’-Ome + 2’-F + PS-ChoL), in MDA-MB-157 and Hs-578T cells. Both siRNA8-mf and siRNA8-mf-PS demonstrated knockdown efficacy comparable to that of unmodified siRNA8 and siRNA10 after 72 h (Fig. 3A, 3B). Notably, effective MYC protein knockdown was observed in MDA-MB-157 cells treated with 20 nM siRNA8-mf-PS-ChoL without the use of a transfection reagent, highlighting the potential of ChoL conjugation as a delivery system (Fig. 3B).
Further investigation of the long-term knockdown efficacy revealed that the optimal duration ranged from 3 to 9 days or more (Fig. 3C, Supplementary Fig. 2A). This sustained efficacy, along with enhanced potency, was more pronounced in cells treated with siRNA8-mf and siRNA8-mf-PS (Fig. 3C-3E). Specifically, siRNA8-mf exhibited significantly greater knockdown efficacy than siRNA8 on day 9 (
The
Additionally, the serum stability of the modified siRNAs siRNA8-mf and siRNA8-mf-PS was compared to that of unmodified siRNA8 and siRNA10. The modified siRNAs showed prolonged stability
A major drawback of siRNAs is the frequent occurrence of off-target effects. The primary objective in designing the asiRNAs was to reduce seed-mediated off-target effects from the seed region (2-8 bases at the 5’ end). Among the designed asiRNAs, those with greater than 60% knockdown efficiency and at least 3 nucleotides removed at the 5’ end of the passenger strands were selected for further optimization through various modifications. With the exception of 19+2/15+2, 19+2/14+2, 18+2/15+0 and 18+2/15+2 (with dTdT), all other designs, including 19+2/18+2, 19+2/17+2, 19+2/16+2, 19+2/16+0, 19+2/15+0 and 19+2/13+2, showed similar (
Among the asiRNAs used in this study, 19+2/16+2 (asiRNA10-3) and 19+2/13+2 (asiRNA10-6) were selected for further optimization and implementation of different modification patterns to enhance knockdown efficacy. Full modification of asiRNA10-3 with 2’-Ome, 2’-F, PS and VP enhanced the knockdown efficiency compared to that of the unmodified siRNAs (Fig. 4F, 4G). Compared with unmodified asiRNA10-3, fully modified asiRNA-VP increased the knockdown efficacy by 19% (Fig. 4F, 4G) and amplified the potency by 7.7-fold (Fig. 4J). MYC protein knockdown efficacy was greater in cells treated with 1 nM asiRNA-VP than in those treated with 20 nM unmodified asiRNA10-3 (Fig. 4G). This difference was also reflected in the IC50 prediction results (Fig. 4J). The knockdown efficacy of asiRNA-VP remained robust even at a lower dose (1 nM). Moreover, compared with unmodified asiRNA10-3, asiRNA-VP not only demonstrated increased potency but also induced enhanced cell death (Fig. 4H, 4I). Additionally, scramble and mismatch control siRNAs, designed from the parent siRNA10, were validated for their lack of effect on mRNA and protein expression, as well as cell viability. Neither the scramble nor the mismatch control siRNA significantly decreased (
Alternate modifications involving equal proportions of 2’-Ome and 2’-F may not always yield beneficial outcomes. For example, the 2’-F modification tends to be less stable than 2’-Ome. Additionally, antisense oligonucleotides modified with 2’-F have shown greater affinity for binding to cellular proteins than those modified with 2’-Ome, leading to interference with protein folding due to increased hydrophobicity, which can result in toxicity (Shen
Knockdown of MYC by high doses of heavily modified siRNAs, such as Mod_asiRNA10-6 and Mod_asiRNA12-1, in the MDA-MB-231 cell line induced either MYC overexpression or an unperturbed response when treated with siRNA8-mf-PS. Moreover, MYC overexpression was observed in MDA-MB-231 cells treated with asiRNA-VP (Supplementary Fig. 5A). We aimed to investigate differences in siRNA sequence, dose and modification type rather than the target genes affected. Various unmodified and modified siRNAs, including siRNA10, asiRNA10-3, asiRNA-VP (at different concentrations), siRNA8, siRNA8-mf, and siRNA8-mf-PS, were tested to determine whether they could induce
To investigate the
The results showed efficient knockdown of MYC mRNA and protein in the mouse xenograft model treated with a single dose of asiRNA-VP-DCA (Fig. 6B, 6C). However, variability in response to treatment was evident, affecting both mRNA and protein levels. In the group receiving four repeated doses of asiRNA-VP-DCA, either SC or IT, knockdown of the MYC protein was still detectable after 46 days of treatment (Fig. 6D), confirming the sustained efficacy of fully modified asiRNA-VP-DCA
In this study, we designed siRNAs targeting the
The mRNA expression levels initially decreased following treatment with unmodified siRNAs and those modified with 2’-Ome or 2’-F but began to increase after 9 days. In contrast, siRNAs modified with PS, specifically siRNA8-mf-PS, maintained their knockdown efficacy longer, demonstrating a slower loss of effect than siRNA8 and siRNA8-mf (Fig. 3C). The prolonged activity of PS-modified siRNAs can be attributed to the increased protein binding affinity conferred by PS modification, which enhances intracellular trafficking and overall potency (Crooke
Serum stability studies further underscored the benefits of siRNA modifications. Compared with their unmodified counterparts, siRNAs modified with 2’-Ome and 2’-F and PS at both 5’ and 3’ ends of the sense and antisense strands exhibited greater stability when incubated with 100% fetal bovine serum (FBS) at 37°C (Fig. 3G, 3H, Supplementary Fig. 3A-3D). Unmodified siRNA8 and siRNA10 were quickly degraded, showing an 80% reduction in just 2 h, with complete degradation occurring within 6 h (Supplementary Fig. 3A, 3D). Conversely, siRNA8-mf and siRNA8-mf-PS demonstrated enhanced serum stability, highlighting the protective effect of these modifications (Supplementary Fig. 3B, 3C).
The primary objective of designing asiRNAs was to minimize off-target effects while maintaining the specific targeting capability of the guide strand. By truncating up to 6 nucleotides from the 5’ end of the passenger strand, our results indicated that the knockdown efficacy was largely unaffected (Fig. 4A, 4B, Supplementary Fig. 4A). This strategy is thought to reduce off-target activity by mitigating unintended global gene downregulation effects (Yuan
The seed region of the siRNA can inadvertently affect global gene expression (Biscans
Previous studies have demonstrated that the incorporation of VP substantially enhances the binding affinity of siRNAs for human Argonaute-2 (hAGO2), thereby improving their gene knockdown efficacy both
Previous findings confirmed that reducing the number of 2’-F modifications while increasing the number of 2’-Ome modifications significantly enhances the stability of siRNAs both
Although extensive chemical modifications are critical for enhancing the overall activity of siRNAs, they can sometimes impair the activity of cleavage factor polyribonucleotide kinase subunit 1 (CLP1) (Parmar
Additionally, treatment with asiRNA-VP led to dose-dependent overexpression of
For example, reducing the PS modifications at the 5’ end of the guide strand in Mod_asiRNA8-4 (Fig. 1R) circumvented the overexpression of
The use of DCA conjugation for
In this study, we developed novel asiRNAs, specifically asiRNA-VP and Mod_asiRNA10-6, along with design platforms (19+2/16+2 and 19+2/13+2). These were designed to target the
Authors would like to thank Dr. Na Young Kim and Dr. Hong Seok Choi for their cooperation in the order, procurement and synthesis of asymmetric siRNA from abroad. Ms. Kang Young Im for procuring the reagents and materials used in this study.
This research was funded by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2022R1A6A1A03046247) and ABION BIO.
Authors declare no conflict of interests.
NM: Conceptualization, design, investigation, performing the experiments, data analysis, drafting and overall writing of the manuscript; MS: injected cells and drug to mice, and performed tumor tissue collection; HY: reviewed the initial and final version of manuscript; CC: helped in drafting of manuscripts and reviewed final version of the manuscript; JYC: Engaged in funding acquisition; SH: monitored the design of animal experiment, and reviewed the final version of manuscript; KS: reviewed the initial and final version of the manuscript; YKS: conceptualization, design, overall guidance, funding acquisition, guiding in drafting the initial and final version of manuscript.