Abstract Background Tumor resistance is the primary reason for treatment failure in patients with cancer, while oncolytic viruses (OVs), as a novel therapy, have been rapidly advancing through clinical evaluation and are typically assessed in recurrent tumors that are refractory to standard chemotherapy. However, whether the adaptive process that fosters chemotherapy resistance influences the efficacy of OV therapy is unknown. Methods We analyzed chemo-resistant colorectal cancer (CRC) using in vitro, in vivo, and patient-derived organoid models to assess sensitivity to OVs. Through RNA sequencing analysis and immunohistochemistry were performed in clinical samples that indicated TANK-binding kinase 1 (TBK1) expression. Using single-cell RNA sequencing, flow cytometry, and in vivo neutralization assays to demonstrate that the combination of TBK1 inhibitor (TBK1i) and OVs reprograms the tumor immune microenvironment, particularly by activating natural killer (NK) cells. Through RNA sequencing analysis, we identified intercellular cell adhesion molecule-1 (ICAM-1) as a potential target responsible for NK cell activation. Subsequently, we designed and conducted rescue experiments, both in vitro and in vivo, to validate the influence of ICAM-1 on NK cell activity. Results We demonstrated that chemo-resistant CRC showed decreased sensitivity to the OV in vitro, in vivo, and patient-derived organoids. Further investigation revealed aberrant activation of TBK1 in chemo-resistant CRC, which mediated the activation of the type I interferon pathway and impaired viral replication. TBK1 inhibition enhanced intratumor viral replication and direct oncolysis effect in vitro and augmented the antitumor immunity elicited by OVs in vivo. Immune cell profiles presented that OV/TBK1i combination reshaped the tumor microenvironment and especially activated the NK cell response. Immune cell depletion studies demonstrated that NK cells were required for the synergistic therapeutic activity of the OV/TBK1i combination. Mechanistically, TBK1 inhibition synergized with VSVΔ51 to increase ICAM1 expression in a RIPK1-dependent manner, promoting NK cell-mediated tumor killing. Conclusion This study presents a promising approach for treating chemo-resistant CRC by combining OVs and TBK1i. Keywords: Chemotherapy, Colorectal Cancer, Combination therapy, Natural killer - NK, Oncolytic virus __________________________________________________________________ WHAT IS ALREADY KNOWN ON THIS TOPIC * Oncolytic viruses (OVs) represent a promising therapeutic strategy for cancer treatment via directly lysing tumor cells and driving antitumor immunity. However, the therapeutic efficacy of OVs in clinical applications has been less than expected and remains limited. Investigating combinational strategies and identifying the small molecular compounds that exert synergistic effects with OVs may optimize therapeutic outcomes. WHAT THIS STUDY ADDS * This study indicates that TANK-binding kinase 1 (TBK1) acts as a barrier limiting the efficacy of OVs in chemo-resistant colorectal cancer (CRC). Inhibition of TBK1 enhances the viral replication of OVs in chemo-resistant CRC and activates intercellular cell adhesion molecule 1-mediated natural killer cell immunity, thereby presenting TBK1 inhibitors (TBK1i) as effective sensitizers for OV therapy in the treatment of chemo-resistant CRC. HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY * The findings of this study provide new insights into the resistance to OV therapy and present a promising approach for treating chemo-resistant CRC by combining OVs and TBK1i. Introduction Colorectal cancer (CRC) is the second most prevalent cancer and the third leading cause of cancer-related mortality worldwide.[51]^1 Current treatment strategies encompass chemotherapy, radiotherapy, surgical intervention, and targeted therapies.[52]^2 Clinically, oxaliplatin (OXA) and 5-fluorouracil (5-FU)-based regimens are the front-line chemotherapy approaches for patients with CRC. Despite advancements in treatment, chemotherapy resistance remains a primary cause of therapeutic failure.[53]^3 4 This resistance is typically associated with complex etiologies, including genetic mutations,[54]^5 apoptosis inhibition,[55]^6 tumor microenvironment (TME) changes,[56]^7 and enhanced repair of damaged DNA.[57]^8 Consequently, there is an urgent need for novel therapeutic strategies to combat resistant and metastatic CRC and to induce sustained antitumor immunity, thereby improving patient outcomes. Oncolytic viruses (OVs) represent a promising new approach in cancer therapy. OVs include naturally occurring, attenuated, or genetically engineered viruses that can selectively infect and lyse tumor cells while retaining low toxicity to normal cells.[58]^9 Current approved OVs include H101 (China) engineered adenovirus for head and neck cancer, Talimogene Laherparepvec (T-VEC) engineered herpes simplex virus (HSV-1, USA) for unresectable metastatic melanoma, and DELYTACT (Japan) for glioblastoma. However, clinical application faces multiple challenges: the heterogeneity of tumor tissues and the complexity of cancer cells mean that a single OV may not completely eradicate all cancerous cells, necessitating a combination with other therapies.[59]^10 Furthermore, immune responses triggered by viral invasion can limit their replication capacity.[60]^11 Existing clinical studies demonstrate that while T-VEC combined with pembrolizumab increases the ORR of advanced melanoma to 62%, a subset of patients still experiences limited clinical benefit. This may arise from multifactorial resistance mechanisms, including viral neutralization by host immunity, stromal barriers impeding viral spread, or tumor cell-intrinsic evasion of viral lysis.[61]^12 13 It could undermine the strong safety record that OVs have maintained.[62]^14 15 TANK-binding kinase 1 (TBK1) is a multifunctional signaling protein kinase crucial in regulating immune responses, cell proliferation, and apoptosis, and tumor progression.[63]^16 TBK1 mediates the induction of type I interferons (I-IFNs) (such as IFN-α/β) and antiviral innate immunity via Toll-like receptor (TLR) or Retinoic acid-inducible gene I (RIG-I)-dependent pathways in the innate immune system.[64]^17 18 Increasing evidence[65]19,[66]21 suggests that abnormal activation of the TBK1 kinase is closely linked to cancer development, including in lung, pancreatic, and CRCs. Chien et al[67]^22 discovered that the RalB-Sec5 complex directly recruits and activates the TBK1 kinase signaling pathway, bridging innate immune and cancer signaling. This pathway can inhibit cancer cell apoptosis. However, despite these findings, the relationship between TBK1 and reduced oncolytic sensitivity of OVs in chemo-resistant CRC remains unclear, and the precise roles and molecular mechanisms involved in overcoming chemotherapy resistance need further clarification. Employing small molecules to selectively boost the growth and replication of OVs in tumor sites has shown great potential as an effective strategy.[68]^23 24 In this study, the VSVΔ51 was constructed based on the wild-type VSV by deleting the matrix protein M51 site, which enhances IFN induction capability, significantly reduces toxicity, and retains oncolytic activity. We demonstrate for the first time that TBK1 inhibitors (TBK1i) can recover the effective replication of VSVΔ51 in chemo-resistant CRC and activate intercellular cell adhesion molecule 1 (ICAM1)-mediated natural killer (NK) cell immunity. TBK1 inhibition enhances VSVΔ51-mediated oncolysis and antitumor immunity, offering new insights into OV therapy and providing a novel therapeutic avenue for overcoming tumor chemotherapy resistance. Materials and methods Ethics approval and consent to participate All animal experiments were performed based on the protocol approved by the Institutional Animal Care and Use Committee of Zhujiang Hospital of Southern Medical University (LAEC-2023–068). Patients and samples 10 chemo-responsive and chemo-unresponsive CRC tissues were obtained from the Guangdong Provincial People’s Hospital between 2022 and 2024. The detailed clinical information were shown in [69]online supplemental table S1. Cell culture and virus HCT116, SW620, HCT8, LOVO, MC38, VERO, and HEK-293T were obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). HCT116/OXA, SW620/5FU, HCT8/T, LOVO/5FU, and MC38/OXA were provided by Professor Zhi Shi (Jinan University, Guangzhou, China). All cells were maintained in a medium supplemented with 10% fetal bovine serum (Gibco, USA), 100 U/mL penicillin, and 100 µg/mL streptomycin (Fudebio, China) at 37°C in a 5% humidified CO[2] atmosphere. For detailed information, refer to online supplementary materials. The OV VSVΔ51 was provided by Professor Deyin Guo (Guangzhou National Laboratory, Guangzhou, P R China) and was replicated and amplified in BHK-21 cells. For the production of pseudotyped VSVΔ51ΔG, the backbone of the pseudotyped virus comes from the VSVΔ51 virus, in which the G gene was deleted (VSVΔG). The recombinant VSVΔ51ΔG was recovered in 293 T cells transfected with the plasmid carrying the VSV-G protein. Construct of TBK1/ICAM1 knockout cell lines pLV3-U6-TBK1 (human/mouse)-sgRNA-Cas9-Puro and an empty vector; pLV3-U6-ICAM1 (mouse)-sgRNA-Cas9-Puro and an empty vector from Miao Ling Technology Company (Wuhan, China). HEK-293T cells were transfected with the core vector and packaging plasmids psPAX2 and pMD2.G using polyethyleneimine. After changing the medium 8 hours post-transfection, lentivirus-containing supernatants were collected after 48 hours, and HCT116-OXA/MC38-OXA cells were exposed to the lentivirus. Following this, cells were selected with 2 µg/mL puromycin for 7–10 days to create stable cell lines. LDH cytotoxicity assay Following the manufacturer’s instructions, cell viability was assessed using the lactate dehydrogenase (LDH) Release Assay Kit (Beyotime, China). Cells were plated at a density of 10,000 cells per well in 0.1 mL of medium in 96-well plates. After the treatment, gently collect the supernatant from the cell culture medium. According to the instructions of the LDH assay kit, add an appropriate amount of the supernatant to a 96-well plate, then add the LDH substrate and reaction buffer, and mix well. Incubate at room temperature for 30 min, then add the stop solution to terminate the reaction. Measure the absorbance (Optical Density, OD value) of each well at a wavelength of 490 nm using a spectrophotometer. Each experimental group was set up with three technical replicates and performed with three independent biological repeats. The line graph (bar values) represents the mean values of each experimental group. Quantitative reverse transcription PCR Total RNA was isolated using TRIzol reagent (Invitrogen). Complementary DNA synthesis was performed using a reverse transcription system from Takara. For quantitative reverse transcription PCR, SuperReal PreMix SYBR Green (AG) and an ABI Q5 Detection System were used. The messenger RNA (mRNA) results were normalized to the expression of ACTB. The primer sequences were provided in [70]online supplemental table S2. Plaque assay Briefly, after infection, supernatants or sera-containing viruses were collected and diluted to infect HCT116-OXA/SW620-5FU cells plated in 24-well plates at 90% confluence. After 48 hours of infection, the supernatants were discarded, and the cells were washed multiple times with phosphate-buffered saline (PBS) before adding 0.8% methylcellulose. 48 hours later, the cells were stained with 0.2% crystal violet for 12 hours, and the plaques (plaque forming unit, pfu/mL) were counted. Immunofluorescence HCT116/OXA cells (2×10^5 per dish) were seeded into glass-bottom dishes and incubated with MRT (5 µM) and VSVΔ51 (multiplicities of infection (MOI)=0.001) for 24 hours. Tumor cells were fixed with 4% paraformaldehyde for 15 min at room temperature, rinsed with 1×PBS, and blocked with 0.3% Triton X-100 and 5% Bovine Serum Albumin (BSA) for 30 min at 37°C. Cells were then incubated with p-MLKL or p-RIPK1 antibodies (1:100 or 1:400 dilution) for 1 hour, followed by anti-rabbit Alexa Fluor 555 (1:1,000 dilution). After 4',6-Diamidino-2-Phenylindole (DAPI) staining, dishes were rinsed and analyzed using a Laser Scanning Confocal Microscope, counting foci in 100 cells per condition. Western blot analysis Cells were lysed using the RIPA Lysis Buffer (P0013, Beyotime, Shanghai), and sodium dodecyl sulfate-polyacrylamide gel electrophoresis was performed. Membranes were visualized on a chemiluminescence imaging system (UVITEC, Alliance Q9, UK) using Immobilon Western Chemiluminescent HRP Substrate (Millipore). A certain protein was quantified as the ratio of itself and β-ACTIN/Tubulin intensity bands by ImageJ software. Drugs and animal models This study was approved by the Institutional Animal Care and Use Committee at Zhujiang Hospital of Southern Medical University. The TBK1 inhibitor (GSK8612) was obtained from TargetMOI, while Necrostatin-1 (Nec-1) and ICAM1 inhibitor (A205804) were purchased from MCE. GSK8612 and Nec-1 were dissolved in Dimethyl Sulfoxide (DMSO), with a maximum concentration of 0.001% confirmed as non-toxic to mice. In the subcutaneous xenograft model, dissociated MC38/MC38-OXA cells (1×10^6) in 50 µL PBS were injected subcutaneously into 5-week-old C57BL/6 mice. After 5 days, tumors (∼100 mm³) formed, and mice were assigned to four groups. From days 5 to 9 and 12 to 16, mice received intraperitoneal injections of GSK8612 (10 mg/kg/day) and Nec-1 (1.5 mg/kg/day) for 2 weeks, with intratumoral injections of VSVΔ51 (2×10^6 PFU/day) on days 5 and 9. Tumor length and width were measured every 2 days. After approximately 2 weeks, mice were euthanized, and tumors were extracted and weighed. Tumor volume was calculated as (length×width²)/2. Some tumors underwent single-cell sequencing and flow cytometry, while others were fixed for H&E staining and immunohistochemistry analysis. Multicolor flow cytometry analysis C57BL/6 mice with MC38/OXA subcutaneous tumors were harvested, minced, incubated with a Tumor Dissociation Kit, mouse (Miltenyi, 130–096–730), triturated, passed through a 70 mm screen, resuspended in Fluorescence-Activated Cell Sorting (FACS) buffer, and stained with fluorochrome-conjugated anti-mouse antibodies from BioLegend or eBioscience, as well as appropriate isotype control antibodies. A Zombie Red Fixable Viability Kit (BioLegend) was used to stain dead cells. We followed a “no-wash” sequential staining protocol (BioLegend) to stain dead cells and for surface staining. Intracellular IFN-γ staining followed the IFN-γ intracellular staining protocol (BioLegend). For single-color compensation controls, Ultracomp eBeads (eBioscience) were used and stained with each of nine fluorescently conjugated antibodies according to the manufacturer’s instructions. For the Zombie Red assay, cells from the non-tumor and tumor quadrants, respectively, were used as single-color compensation controls. All samples were run in a BD FACSymphony S6. Data were analyzed with FlowJo software. Technicians acquiring and gating the data were blinded to the treatments. Co-culture experiments HCT116/OXA cells carrying the luciferase reporter gene (HCT116/OXA-luc), were added to 96-well plates in different proportions (2,000, 5,000). On attachment, the cells were treated with either Vehicle, the combination of MRT (5 µM)/VSVΔ51 (MOI=0.001) (Comb), and Nec-1 (20 µM)/ICAM1 inhibitor (100 µM) before combining for 6 hours. Afterward, NK-92 cells were added at 2,000 seeding densities. The luciferase fluorescence signals were analyzed at 0, 6, 12, and 24 hours using the IVIS Lumina system to monitor the co-culture killing effects of HCT116/OXA-luc and NK-92 cells. The results were analyzed by Living Image software. RNA-seq and relative data analysis, Single-cell RNAseq analysis, Construction of human CRC organoids, and Human NK cell sorting were provided in [71]online supplemental file 2. Statistical analysis All statistical analyses were performed using GraphPad Prism software. Comparisons between different groups were made using Student’s t-test or analysis of variance (ANOVA) as appropriate in the in vitro study. Values of tumor volume were analyzed by two-way ANOVA. All error bars indicate SD. Differences were considered significant if the p value was<0.05. Data availability Gene expression data had been deposited in the Gene Expression Omnibus (GEO) repository with the accession code GSE293436. All other data that support the findings of this study were available from the corresponding authors on reasonable request. Results Aberrant activation of TBK1 in chemo-resistant CRC reduces the sensitivity to OVs To explore the therapeutic potential of OVs for combating chemo-resistant CRC, we first tested the oncolytic efficacy of two OV strains, VSVΔ51 and HSV-1, in five chemo-resistant cell lines and their parental CRC cell lines. These cell lines included the OXA-resistant HCT116 (HCT116/OXA), OXA-resistant MC38 (MC38/OXA), 5-FU-resistant SW620 (SW620/5FU), 5-FU-resistant LOVO (LOVO/5FU), and paclitaxel-resistant HCT8 (HCT8/T). CRC cells were treated with varying concentrations of chemotherapeutic agents or different MOI with VSVΔ51 and HSV-1. The sensitivity to different drugs or OVs was quantified using the half-maximal inhibitory concentration (IC[50]). IC[50] measurements showed that the sensitivities to VSVΔ51 and HSV-1 were significantly reduced in chemo-resistant cell lines compared with their parental CRC cell lines via cell counting kit-8 assay ([72]online supplemental figure S1A–D) and LDH release assay ([73]figure 1A,B). Correlation analysis of IC[50] values indicated that the IC[50] values for OVs were positively correlated with those for chemotherapeutic agents, suggesting that chemo-resistant CRC cell lines showed reduced sensitivity to OVs compared with their parental CRC cell lines ([74]online supplemental figure S1E). Figure 1. Aberrant activation of TBK1 in chemo-resistant CRC reduced the sensitivity to OVs. (A–B) The measurement for oncolytic effect of VSVΔ51 and HSV-1 in CRC cell lines. HCT116/OXA, MC38/OXA, SW620/5FU, LOVO/5FU, HCT8/T cells and their parental cells were treated with escalating titers of VSVΔ51 or HSV-1 for 24 hours, then the cell death rate was measured by LDH assay and the sensitivity to VSVΔ51 (A) or HSV-1 (B) was quantified via calculating IC[50] value. (C) C57 mice with MC38 and MC38/OXA subcutaneous tumors were treated with VSVΔ51 (2×10^6 PFU/day, intratumoral injection). (D) Representative images of tumor tissue and tumor weights for each group from D. (E) Tumor volumes were recorded every 2 days (n=5). (F–H) The measurement for the oncolytic effect of VSVΔ51 in CRC organoid. The CRC organoids derived from chemo-responsive patients (n=3) and chemo-unresponsive patients (n=3) were infected with/without VSVΔ51 (MOI=0.01) or untreated for 48 hours. Scale bar=100 µm. #1, patient 1; #2, patient 2. (F) Schematic illustration for the CRC organoid construction model. (G) Representative microscope images of CRC organoid after VSVΔ51 treatment. The images shown represent samples from n=3/6 patients in each group (Chemo-Responsive Group: 3 typical responding cases selected from n=6; Chemo-Unresponsive Group: 3 typical non-responding cases selected from n=6). (H) Cell viability of CRC organoid was detected via measuring the intracellular ATP level. (I) KEGG pathway analysis of RNA-seq data showed the upregulation of IFN-related pathways in HCT116/OXA compared with HCT116 cells. (J) Volcano plot of RNA-seq data showed the significant upregulation of TBK1 in HCT116/OXA cells. (K) Immunoblot analysis of TBK1 in the five pairs of CRC lines. Quantification of p-TBK1 levels represents the ratio of chemo-unresponsive to chemo-responsive cells, normalized to β-actin (mean±SD, n=3 independent experiments): HCT116/OXA (3.387±0.533), SW620/5FU (5.522±0.205), LOVO/5FU (11.390±0.928), HCT8/T (4.042±0.891), MC38/OXA (9.990±0.224). (L) Immunohistochemical analysis of TBK1 expression in clinical CRC patient samples (n=10). Scale bar=50 µm. (M) The bilaterally implanted mice were treated with VSVΔ51 (3×10^7 PFU/day) by intravenous injection. MC38 and MC38/OXA cells were implanted at the left side (L) and right side (R) of mice. Tumors were resected 24 hours post-injection. Viral titers of subcutaneous tumors were detected by plaque assay. (N) Immunofluorescence staining was used to evaluate the expression of GFP (reporter gene for VSVΔ51). Representative images of n=5. Scale bar=100 µm. Statistical significance was determined using one-way ANOVA in A, B, two-way ANOVA in E, F, or two-tailed Student’s t-test in M, N. Data represent the mean±SD. n=3 biological replicates in A, B, L. n=5 biological replicates in E, F, N. *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001; n.s., non-significant. ANOVA, analysis of variance; CRC, colorectal cancer; 5-FU, 5-fluorouracil; HSV-1, herpes simplex virus 1; IC[50], half-maximal inhibitory concentration; LDH, lactate dehydrogenase; MOI, multiplicities of infection; OV, oncolytic virus; OXA, oxaliplatin; TBK1, TANK-binding kinase 1; ATP, adenosine triphosphate; JAK-STAT, janus kinase–signal transducer and activator of transduction pathway; PFU, plaque forming unit; KEGG, kyoto encyclopedia of genes and genomes. [75]Figure 1 [76]Open in a new tab To confirm the above findings, we next tested the oncolytic effects of VSVΔ51 in vivo and patient-derived tumor organoids. For the in vivo assays, we established subcutaneous tumors using MC38 or MC38/OXA cells in immune-competent C57BL/6 mice ([77]figure 1C). We found that VSVΔ51 treatment via intratumoral injection significantly inhibited tumor growth and tumor burden in MC38 subcutaneous tumors, but not in MC38/OXA subcutaneous tumors ([78]figure 1D,E). Furthermore, we collected three pairs of chemo-responsive and chemo-unresponsive tumor organoids derived from patients with CRC to evaluate the oncolytic effect of VSVΔ51 ([79]figure 1F). Time-lapse microscopic examination revealed that VSVΔ51 treatment triggered tumor sphere lysis in chemo-responsive organoids; however, chemo-unresponsive organoids showed no significant morphological changes under VSVΔ51 treatment ([80]figure 1G and [81]online supplemental figure S1F). The intracellular ATP measurement confirmed that VSVΔ51 treatment did not significantly reduce the cell viability of chemo-unresponsive organoids ([82]figure 1H). These data demonstrated that the sensitivity of CRC tumors to OVs was decreased during the acquisition of chemo-resistance. To investigate the underlying mechanisms, RNA sequencing (RNA-Seq) was conducted in HCT116 and HCT116/OXA cells without stimulation. KEGG pathway enrichment analysis revealed that HCT116/OXA cells upregulated the genes relating to I-IFN signaling pathway and JAK-STAT cascade compared with HCT116 cells ([83]figure 1I). Strikingly, the mRNA level of TBK1, a critical kinase in the I-IFN signaling pathway, was upregulated in HCT116/OXA cells ([84]figure 1J). We further evaluated the mRNA expression of genes related to the I-IFN pathway at virus-free conditions and found that both HCT116/OXA and SW620/5FU cells showed higher expression of IFNB1, ISG15, and CXCL10 compared with their parental CRC cell lines ([85]online supplemental figure S1G). Western blot analysis of five paired CRC cell lines also confirmed the upregulation of the protein level of TBK1 and the phosphorylation of TBK1 and IRF3 in chemo-resistant cells ([86]figure 1K). To clinically validate the upregulation of TBK1, immunohistochemistry was performed in clinical samples from 10 paired chemo-responsive and chemo-unresponsive patients with CRC ([87]online supplemental table S1). The results indicated TBK1 overexpression in the chemo-unresponsive samples ([88]figure 1L). Considering the pivotal role of TBK1 in antiviral immunity, we next evaluated the viral replication of OVs in chemo-resistant CRC tumors in vivo. A bilateral tumor model was established to analyze the viral replication and tropism of VSVΔ51 ([89]figure 1M). Viral titer detection and immunofluorescence analyses demonstrated that MC38/OXA tumors showed the reduction of virus replication of VSVΔ51 in vivo, compared with MC38 tumors ([90]figure 1N). The above data suggested that the aberrant activation of TBK1 led to the reduced oncolysis and sensitivity of OVs in chemo-resistant CRC. TBK1 inhibition potentiates oncolytic efficacy of VSVΔ51 in chemo-resistant CRC The aberrant activation of TBK1 contributes to the proliferation and survival of tumor cells, particularly in specific mutational or tumorous contexts.[91]^25 26 TBK1 is also identified as an immune-evasion gene, and targeting TBK1 can enhance the response to PD-1 blockade by sensitizing tumor cells to effector-cytokine-induced cell death.[92]^27 These findings drove us to test whether the combination of TBK1i could overcome the resistance to OV in chemo-resistant CRC. We conducted a combination treatment using VSVΔ51 and MRT67307/GSK8612, a classical inhibitor of TBK1, in the HCT116/OXA and SW620/5FU cell lines. Cell viability was measured after treatment with increasing doses of MRT67307/GSK8612 in the presence or absence of VSVΔ51. Differences in the area under the curve (DAUC) were calculated based on the presence or absence of VSVΔ51. The results demonstrated that MRT67307/GSK8612 treatment significantly enhanced the sensitivity of VSVΔ51 (MRT67307 DAUC=0.73 for HCT116/OXA and 0.51 for SW620/5FU; GSK8612 DAUC=0.53 for HCT116/OXA and 0.35 for SW620/5FU) ([93]figure 2A and [94]online supplemental figure S2A). We further quantified this sensitization effect by measuring changes in the IC50 shift. Treatment with MRT67307/GSK8612 sensitized the chemo-resistant CRC to VSVΔ51 to varying degrees, with an IC50 shift of MRT67307 being 3,008-fold for HCT116/OXA and 3,029-fold for SW620/5FU, GSK8612 being 1,059-fold for HCT116/OXA and 266-fold for SW620/5FU ([95]figure 2B and [96]online supplemental figure S2B). Fluorescence imaging and flow cytometry results revealed that MRT67307/GSK8612 treatment facilitated the replication of VSVΔ51 in the HCT116/OXA and SW620/5FU cell lines ([97]figure 2C and [98]online supplemental figure S2C). Furthermore, a single treatment of VSVΔ51 or MRT67307/VSVΔ51 combination can exert a cytotoxic effect in chemo-sensitive HCT116 and SW620 cell lines ([99]online supplemental figure S2D). To exclude the off-target effects of the TBK1 chemical inhibitor, we performed CRISPR–Cas9 knockout using two different single-guide RNAs targeting TBK1 and confirmed the successful knockout through immunoblot ([100]online supplemental figure S2E). Knockout of TBK1 significantly increased the oncolytic effect of VSVΔ51 in HCT116/OXA and MC38/OXA cells ([101]figure 2D and [102]online supplemental figure S2F). Viral titer assay confirmed the enhancement of VSVΔ51 production following MRT67307 treatment ([103]figure 2E). In chemo-unresponsive CRC organoids, the combination treatment of MRT67307 and VSVΔ51 also induced severe lysis of tumor spheres and reduced cell viability ([104]figure 2F). Figure 2. TBK1 inhibition potentiated the sensitivity of chemo-resistant CRC to VSVΔ51 in vitro and in vivo. (A) HCT116/OXA and SW620/5FU cells were treated with increasing doses of the TBK1 inhibitor MRT67307 (MRT) for 24 hours, either in the absence or presence of VSVΔ51 (MOI=0.001), then the cell viability was assessed. The differences in the area under the curve (DAUC) were calculated according to the formula (AUC[single]–AUC[combined])/AUC[combined]; orange areas represent DAUC. (B) IC[50] shifts were determined treated with escalating titers of VSVΔ51, with or without MRT (5 µM) for 24 hours. (C) MRT treatment promoted the viral infection of VSVΔ51. Cells were infected with VSVΔ51 (MOI=0.001) in the presence or absence of MRT (5 µM). The fluorescence imaging was monitored for viral reporter GFP. Scale bar=100 µm. The ratio of infected cells (GFP^+) was measured by flow cytometry. (D) TBK1 knockout was performed in HCT116/OXA cells by CRISPR–Cas9 system using different sgRNAs. Cells were infected with VSVΔ51 (MOI=0.001) for 24 hours, then cell viability was measured by CCK-8. (E) Plaque assays were conducted to evaluate the titer of VSVΔ51. Cells were infected with VSVΔ51 (MOI=0.001) in the presence or absence of MRT (5 µM). Then the cellular supernatant was harvested for the plaque assay. (F) MRT treatment enhanced the oncolytic effect of VSVΔ51 in chemo-unresponsive CRC organoids. The viability of CRC organoids was monitored at 48 hours after treatment of VSVΔ51 and MRT via measuring the intracellular ATP. Scale bar=200 µm. (G) Nude mice with HCT116/OXA subcutaneous tumors were treated with TBK1 inhibitor GSK8612 (10 mg/kg/day, intraperitoneal injection), VSVΔ51 (2×10^6 PFU/days, intratumoral injection), or a combination treatment. (H) Representative tumor images for each group from G. Tumor volume and weight were presented as the mean±SD (n=5). (I) MC38/OXA subcutaneous xenografts in C57 mice were treated with GSK8612 (10 mg/kg/day, intraperitoneal injection), VSVΔ51 (2×10^6 PFU/days, intratumoral injection), or a combination treatment. (J) Representative images, weights, and tumor volume for each group mentioned in I. Tumor weight and volume were presented as the mean±SD (n=5). (K) Tumor size was recorded every 2 days and the tumor-free ratio was calculated at the end of the experiment. (L) Viral gene VSV-G was measured by qPCR in C57 mice tumors. Statistical significance was determined using one-way ANOVA in A, B, F, two-way ANOVA in H, J, K, or two-tailed Student’s t-test in D, E, L. Data represent the mean±SD. n=3 biological replicates in A, B, C, D, E and F. n=5 biological replicates in H, J, K and L. *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001; n.s., non-significant. ANOVA, analysis of variance; CRC, colorectal cancer; 5-FU, 5-fluorouracil; IC[50], half-maximal inhibitory concentration; MOI, multiplicities of infection; mRNA, messenger RNA; OXA, oxaliplatin; PBS, phosphate-buffered saline; qPCR, quantitative PCR; sgRNA, single-guide RNA; TBK1, TANK-binding kinase 1; ATP, adenosine triphosphate; PFU, plaque forming unit. [105]Figure 2 [106]Open in a new tab We next evaluated the therapeutic potential of the VSVΔ51/TBK1i combination in vivo. GSK8612 is a specific inhibitor of TBK1 and is suitable for assessing in vivo. Nude mice with HCT116/OXA subcutaneous tumors were randomized into four groups and treated with (1) vehicle, (2) intraperitoneal injection of GSK8612, (3) intratumoral injection of VSVΔ51, and (4) a combination of GSK8612 and VSVΔ51 ([107]figure 2G). The VSVΔ51/GSK8612 combination treatment markedly inhibited tumor growth and burden ([108]figure 2H). Next, we developed subcutaneous tumors using MC38/OXA cells in immunocompetent C57BL/6 mice ([109]figure 2I). The VSVΔ51/GSK8612 combination treatment induced tumor regression in the mice and, at the same time, led to the complete tumor elimination in one of five mice ([110]figure 2J,K), indicating that the VSVΔ51/GSK8612 combination contributed to antitumor immunity. Meanwhile, GSK8612 treatment significantly elevated VSV-G expression in tumor tissues but not in the heart, liver, spleen, lungs, and kidneys ([111]figure 2L and [112]online supplemental figure S2G,H). Furthermore, this combination therapy exhibited no significant toxicity to major organs such as the heart, liver, spleen, lungs, and kidneys, indicating a favorable biosafety profile ([113]online supplemental figure S2I). These data suggest that TBK1 inhibition facilitates oncolysis by amplifying viral replication and enhancing antitumor immunity. VSVΔ51/TBK1i combination reshapes the immune profiles and enhances natural killer cell response The remarkable therapeutic effect observed in the immune-competent model prompted us to investigate the impact of the VSVΔ51/GSK8612 combination on the immune profile. We conducted single-cell RNA sequencing (scRNA-seq) analysis on CD45^+ cells isolated from MC38/OXA tumors in C57BL/6 mice treated with either TBK1i, VSVΔ51, or the VSVΔ51/TBK1i combination ([114]figure 3A). After standard data processing and quality control procedures, we obtained transcriptomic profiles for 90,651 cells. Unsupervised clustering identified 19 distinct clusters ([115]figure 3B and [116]online supplemental figure S3A). We further classified these clusters into 10 cell types by canonical cell type specific markers, including T cells, monocytes/macrophages, neutrophils, basophils, dendritic cells (DCs), plasmacytoid DCs, NK cells, B cells, fibroblasts, and erythrocytes ([117]figure 3C,D). By quantifying the cell composition of the samples, we found that combination treatment increased the frequency of macrophages and NK cells; unexpectedly, the frequency of T cells decreased in the combination group ([118]figure 3E). Figure 3. TBK1 inhibitor combined with VSVΔ51 remodels the tumor immune microenvironment. (A) Single-cell sequencing workflow diagram. (B) The two-dimensional plots of UMAP dimensionality reduction for cell type. (C) Dot plot showing marker genes for 10 cell types. (D) The two-dimensional plots of UMAP dimensionality reduction for 10 clusters. (E) The percentage of 10 clusters between each sample was represented on a proportion chart. (F–H) UMAP of reclustered macrophages, showing changes in the proportions of various subtypes between different groups. (I–K) UMAP of reclustered T and NK cells, showing changes in the proportions of various subtypes between different groups. (L) Violin plots show the differential expression of immune-related genes among subtypes of T/NK cells. (M) Volcano plot shows that the expression of Gzmc in the combination group was significantly higher than in the other groups. n=2 biological replicates. DC, dendritic cell; NK, natural killer; pDC, plasmacytoid dendritic cell; scRNA-seq, single-cell RNA sequencing; TBK1, TANK-binding kinase 1; UMAP, uniform manifold approximation and projection. [119]Figure 3 [120]Open in a new tab We then reanalyzed macrophages and got six functional subclusters ([121]figure 3F and [122]online supplemental figure S3B). The composition of subclusters in each treatment group showed no significant differences; however, the proportion of the C1_C1qa subcluster was significantly increased in the combination treatment group ([123]figure 3G,H). Studies have shown that C1q^+ macrophages express multiple inhibitory molecules, and their high infiltration was significantly correlated with poor prognosis.[124]^28 We continued to analyze T and NK cells, which were divided into 14 subgroups with distinguishable signatures ([125]figure 3I and [126]online supplemental figure S3C). Based on the internal statistical proportions of the samples, the cell proportions of CD4T_C1_Treg, CD8T_C4_Gzmf, CD8T_C5_Ifng, and NK_C1_Ccl5 were elevated in the combination or GSK groups, with a much higher proportion of NK_C2_Gzmc in combination groups ([127]figure 3J). The increase in NK subclusters was also observed in the T/NK group preferences by Ratio of observed to expected