Introduction

GRB2 is an intracellular adapter protein essential for cell proliferation that consists of a central SH2 domain flanked by two SH3 domains1,2,3,4,5. Classically its SH3 domains direct complex formation with proline-rich regions of other proteins, and its SH2 domain binds tyrosine phosphorylated sequences6,7,8,9,10. Cytoplasmic GRB2 acts in initial steps of receptor tyrosine kinase (RTK) signaling to the Ras-MAPK cascade. In the nucleus GRB2 adapter moonlights in initial steps for efficient homology-directed-DNA of DNA double strand breaks (DSBs)11. Given these dual functions, we reasoned that GRB2 could act more generally in the DNA damage response (DDR) including its DNA replication stress responses and the activation of the innate immune response by loss of genome stability during replication. In particular, tumor cells with oncogenic replication stress might select for GRB2 activities associated with proliferation and the DDR. Yet GRB2 nuclear activities and mechanisms as well as their possible connections to cancer are largely undefined.

Here we tested and defined GRB2 interactions and activities with DNA replication fork proteins in vitro, in cells, and in vivo. We find that GRB2-depleted cells strikingly mirrored DNA replication fork protection characteristics of the BRCA2-deficient phenotype. In response to replication stress from hydroxyurea (HU) and PARP inhibitor (PARPi), GRB2 stabilized RAD51 on single-stranded DNA (ssDNA) at stalled replication forks to reduce cytoplasmic DNA accumulation and cGAS/STING activation. Furthermore, BRCA2 knockdown (KD) in a GRB2-KO background showed no added fork degradation defects consistent with an epistatic relationship. During RF stress, GRB2 depletion promoted cGAS/STING activation that translated into the production of inflammatory cytokines and recruitment of cytotoxic T-cells. In a GRB2-depleted syngeneic ovarian cancer mouse model, PARPi treatment led to enhanced targeted destruction of tumor cells by the host immune system compared to PARPi alone. These findings align GRB2 with BRCA2 for replication fork protection and PARPi responses that include the therapeutic activation of innate immunity.

Results

GRB2 binds DNA replication forks and functions in the replication stress response

GRB2 in the cytoplasm acts in growth factor receptor tyrosine kinase (RTK) and Ras/MAP kinase activation driving quiescent (G0) cells to enter cell-cycle. Consistent with these observations, our previously generated GRB2 knockdown (GRB2-KD) in A431 cells12 showed a measurable G1-phase stagnation and reduced S-phase duration compared with wild type (control) without affecting overall cell viability. We therefore asked whether nuclear GRB2 may impact efficient S-phase progression. We treated cells with low dose hydroxyurea (HU) to create replication stress and found that GRB2-KD cells showed a dramatic increase in S-phase stagnation within a 24 h time period (Supplementary Fig. 1a). GRB2-KD cells, challenged with HU, required a longer time to complete DNA duplication implying GRB2 acts in maintaining S-phase efficiency under replication stress. Similar results were observed in GRB2 knockout (KO) HAP-1 cells (Supplementary Fig. 1b). A GRB2 functional role was also shown in colony survivals assay where HU and mitomycin C (MMC) treatment severely impeded colony formation in GRB2-KO cells that can however be rescued by reconstitution of GRB2 (Supplementary Fig. 1c, d).

Mass spectrometry previously identified proliferating cell nuclear antigen (PCNA) as a nuclear GRB2 interacting protein11. PCNA a trimeric docking protein for recruitment of replication fork (RF) proteins including DNA polymerases, FEN1 nuclease, and DNA ligase13. We therefore investigated GRB2 at the RF using fluorescence lifetime imaging microscopy (FLIM) to measure fluorescence resonance energy transfer (FRET) between GRB2 and PCNA. Under normal conditions GRB2 co-localized with PCNA, but overlap was difficult to assess (Fig. 1a, left panel and Supplementary Fig. 2a). Yet, FLIM showed a measurable FRET between PCNA and GRB2 indicated by the reduction in the average lifetime of the GFP-tagged PCNA (Fig. 1a, right panel and Supplementary Fig. 2b). mEmerald-PCNA overexpressed cells showed some cytoplasmic localization when co-expressed with RFP-tagged GRB2. Investigation of potential GRB2-meditated PCNA retention in the cytoplasm found that cytoplasmic-PCNA localization is likely an overexpression artifact (Supplementary Fig. 2c). Indeed, FLIM accurately measures interaction between GRB2 and PCNA inside the nucleus circumventing contrast-based imaging localization artifacts (Supplementary Fig. 2a, b). The direct interaction between PCNA and GRB2 was further supported by an in vitro binding assay using purified proteins (Supplementary Fig. 2c, d). We then identified residue 200-209 (QTGMFPRNY) as an embedded putative PCNA interaction peptide (PIP) motif within the C-terminus of GRB2. We found that synthetic GQTGMFPRNY peptide bound PCNA with a dissociation constant comparable to WTGRB2 (Supplementary Fig. 1c). As PCNA is tyrosine phosphorylated on residue Y21114,15,16 to form a potential pYxNx GRB2 SH2 domain binding site, we tested binding and found that tyrosine phosphorylated PCNA (pPCNA) binds GRB2 with an order of a magnitude higher affinity in the MST binding assay (Supplementary Fig. 2c). Given the close association of PCNA with the DNA replication fork (RF), these collective data supported GRB2 interactions and function at the RF.

Fig. 1: GRB2 at the DNA replication fork inhibits MRE11 mediated fork-degradation.
figure 1

a FLIM/FRET showing GRB2-PCNA direct interaction within an apparent PCNA replication focus. Colocalization shown in the left, while false colored lifetime image on the right generated by pixel-by-pixel map** of the measured lifetime-values represented by the scale 1.7–2.2 nanoseconds. Scale bar 25 μm (b) iPOND assay showing GRB2 association with replication DNA, and enriched on the nascent DNA in response replication stress. PCNA was used as a positive control. c Enhanced fork degradation in GRB2-depleted HeLa cells under replication stress. d An independent set of experiments with GRB2 reconstitution. Only WTGRB2 (KO + GRB2), but not the K109R mutant alleviated replication stress induced fork degradation in GRB2-depleted HeLa cells. Radiometric analysis of ≥200 fibers, n = 3. e Replication stress induced increased ssDNA in GRB2-depleted cells under replication stress indicating enhanced nuclease activity in HeLa cells. Scale bar 10 μm. f Quantitation of the ssDNA intensities from three independent experiments represented in (e), intensities of 100–300 foci, n = 3. g Fork degradation is inhibited by MRE11 nuclease inhibitor Mirin in HeLa cells lacking GRB2 and under replication stress. h MRE11knockddown prevents fork degradation in GRB2-deplered HeLa cells under replication stress. Fiber assay radiometric analysis of ≥ 200 fibers, n = 3 in (c, d, fh). The significance was analyzed by two-sided Student’s t test. ***P ≤ 0.001, and ****P ≤ 0.0001; NS not significant. Error bars showing standard deviations (SD). For (bd) and (fh), source data are provided as a Source Data file.

We therefore investigated GRB2’s direct association with the RF by using the isolation of Proteins On Nascent DNA (iPOND) method17,18. This assay was performed in cells unperturbed or challenged with HU-induced replication stress. As expected, GRB2 was present in the progressing RFs marked with PCNA accumulation. Interestingly, more GRB2 was loaded onto the stalled RF, despite the apparent PCNA dissociation under HU treatment, suggesting other potential GRB2 interactions at the RF (Fig. 1b). This enrichment of GRB2 on nascent DNA at stalled replication forks was validated by the quantitative in situ analysis of protein interactions at DNA replication forks (SIRF) assay (Supplementary Fig. 3)19.

We further investigated the GRB2 function at RFs with DNA-fiber assay measurements of replication fork progression between control and GRB-KO cells (Supplementary Fig. 4a–c). IdU track length measurements under normal cell growth conditions found no significant difference between the WT and GRB2-KO HeLa and HAP-1 cells (Fig. 1c and Supplementary Fig. 4c,d). However, with replication stress, a significant reduction in IdU stained DNA tract led to decreased IdU/CIdU ratio in GRB2-KO cells (Fig. 1c and Supplementary Fig. 4d). Importantly, GRB2-KO cells reconstituted with exogenous GRB2 restored replication progression to levels comparable to wild-type parental cells (Fig. 1d). These data show GRB2 localizes to RFs and functions in the replication stress response.

GRB2 protects stalled replication forks from MRE11-mediated degradation

As mutant GRB2K109R disrupts the GRB2-MRE11 interaction for DNA double-strand break repair while retaining RTK activity11, we employed the impact of this separation-of-function mutant on stalled RFs. We found that reconstitution with GRB2K109R failed to rescue fork degradation in GRB2-KO cells (Fig. 1d). This finding is consistent with RF degradation by MRE11 in GRB2-KO cells and suggested GRB2 interaction with MRE11 can restore RF protection.

Unprotected stalled RFs can undergo extensive nucleolytic processing generating single stranded (ss) DNA20,21. We therefore employed immunofluorescence analysis with an antibody to single-stranded (ss) DNA (ssDNA). We measured a significant increase in ssDNA accumulation in GRB2-KO cells treated with HU compared to control cells (Figs. 1e, f). We then investigated if the observed reduction in IdU stained DNA tract and the accumulation of ssDNA was the result of MRE11 nuclease processing of stalled RFs. Specifically, we employed the single-molecule DNA fiber assay to investigate the effect of MRE11 inhibitor Mirin on HU-induced fork degradation. Mirin treatment had no effect on the integrity of stalled RFs in the control HeLa or HAP1 cells, while in GRB2-KO cells HU-induced fork degradation was prevented (Fig. 1g and Supplementary Fig. 4e). The specificity of MRE11-mediated stalled replication fork processing was further confirmed by MRE11-knockdown (KD), which like Mirin rescued fork degradation in GRB2-KO HeLa cells (Fig. 1h and Supplementary Fig. 5a). We also examined other RF processing nucleases, EXO1 and DNA2. Knockdown of EXO1 or DNA2 failed to rescue fork degradation in GRB2-KO cells (Supplementary Fig. 5b).

These collective data indicate GRB2-MRE11 interaction blocks excessive processing of stalled replication forks to limit generation of ssDNA. This is supported by the findings that GRB2-MRE11 interaction disrupting K109RGRB2 mutant failed to prevent MRE11-mediated stalled RF degradation (Fig. 1d) or to restore cell replication stress resistance induced by HU or MMC treatment in colony survival assays (Supplementary Fig. 1c).

GRB2 depletion mirrors BRCA deficiency in loss of replication fork protection

In BRCA deficient cells, MRE11-dependent fork degradation creates long stretches of ssDNA, as one of the main causes of PARP inhibitor sensitivity22. Our GRB2 KO HeLa and HAP-1 cells express BRCA2

Fig. 3: GRB2 promotes reverse fork stability by inhibiting RAD51 ATPase activity.
figure 3

a RAD51 interacts with the SH2 domain of GRB2. MST binding isotherms measuring binding affinity of RAD51 with wild type GRB2, SH2 domain and indicated GRB2 mutant. The binding affinities (Kds) mean values with ±SD are shown below, n = 3. b ATPase activity assay showing GRB2 dose-dependent inhibition of RAD51 ATP-hydrolysis with ±SD, n = 3. c Representative gel-image showing GRB2 dose dependent RAD51 strand-stabilization to dsDNA with 5′ overhang. d Normalized quantitation of RAD51 strand exchange and stability induced by GRB2 with ±SD, n = 3. e Representative PLA images of RAD51 foci formation with ssDNA. f Quantitation of nascent DNA associated RAD51 collected from >350, n = 3. Scale bar 10 μm. The significance was analyzed by two-sided Student’s t test. ****P ≤ 0.0001; NS not significant. For ad and f source data are provided as a Source Data file.

ATP hydrolysis releases RAD51 from ssDNA40,41, and ATPase-deficient RAD51 mutant protects RFs from degradation in GRB2-KO cells. We therefore asked if GRB2 could protect RFs by inhibiting RAD51 and tested the effect of GRB2-RAD51 interaction on RAD51 ATPase activity. GRB2 was titrated into a fixed concentration of RAD51, and ATPase activity was measured. The results showed a dose-dependent inhibition of RAD51 ATPase activity with increasing concentration of GRB2 (Fig. 3b). The inhibition of RAD51 ATP hydrolysis in vitro was shown to promote stable strand exchange and formation of a stable complex with dsDNA with a 5′ overhang41. We therefore mixed a 32nt duplex DNA with one strand fluorescently labeled with a 90nt unlabeled ssDNA containing complementary sequence to the labeled 32nt and monitored the exchange of the labeled 32nt. The results showed that GRB2, in a dose-dependent manner, induced an efficient RAD51-mediated exchange of labeled 32nt with the complementary 90nt sequence (Fig. 3c, d).

To further test the impact of GRB2 on RAD51 stability at the replication fork, we used the proximity ligation SIRF assay19 to measure RAD51 proximity to nascent ssDNA. In cells under normal growth condition, there was no significant difference in RAD51 localization between the control (WT) and GRB2-KO (KO) cells (Fig. 3e). In WT cells, HU treatment induced a significant increase in RAD51-bound nascent DNA foci compared to GRB2-KO cells. HU treated GRB2-KO cells showed a significantly lower level of DNA-bound RAD51 (Fig. 3e, f), while the expression pattern of RAD51 was not affected by the GRB2 Knockout (Supplementary Fig. 6f). These data are consistent with in vitro observations where higher RAD51 ATPase activity correlated with decreased strand stabilization to 5′-overhang dsDNA without GRB2 (Fig. 3b–d). The RAD51 K133R mutant is ATP-hydrolysis deficient25,27,36,37. To further understand the impact of GRB2 on RAD51 ATPase activity and replication fork stability, we performed dose-escalation overexpression analysis of the ATPase-deficient K133R-RAD51 in GRB2-KO cells. In control cells with normal levels of GRB2, K133R expression level had little impact. However,  a 2-fold higher overexpression of K133R in GRB2-KO cells was sufficient to rescue PARPi sensitivity to a level comparable to the parental control cells (Supplementary Fig. 6h, i). Thus, GRB2 evidently protects RFs by inhibiting RAD51 ATPase activity to maintain stable RAD51-DNA interactions. We envisage that in the absence of GRB2, ATP hydrolysis of RAD51 loaded onto reversed fork leads to its premature dissociation from DNA, leaving them vulnerable to MRE11-dependent nucleolytic processing.

GRB2 restricts cGAS/STING activation and inflammatory cytokine release under replication stress

In general, PARPi sensitivity in BRCA1/2 defective cells and tumors correlates with fork protection defects that can generate cytosolic ssDNA and trigger cGAS/STING activation21,42,43,44. We therefore examined cells with GRB2 deficiency and found that they are vulnerable to MRE11-medidated degradation of reversed RFs, and therefore sensitive to replication stress induced by HU or MMC treatment (Supplementary Fig. 1c, d). GRB2-KO cells are also sensitive to PARPi11, and PARPi sensitivity induces cytoplasmic micronuclei formation26,45,46. We therefore tested and found PARPi (Olaparib) treatment induced a significantly higher level of cytoplasmic micronuclei in GRB2-depleted HAP-1 and HeLa cells compared to controls (Fig. 4a and Supplementary Fig. 7a, b). Notably, reconstitution of GRB2 in GRB2-KO cells suppressed Olaparib-induced micronuclei (Fig. 4b and Supplementary Fig. 7a, b) underscoring the importance of GRB2 levels in control of micronuclei.

Fig. 4: PARPi induced DNA replication stress in GRB2 depleted cells cause genomic instability, cytoplasmic DNA accumulation resulting in cGAS/STING activation and chemoattractant secretions.
figure 4

a Representative PicoGreen and DAPI staining after 10 μM Olaparib treatment for 48 h and the resulting cytoplasmic micronuclei formation in HAP-1 cells. Scale bar 10 μm (b) Quantitation of cytoplasmic micronuclei form three independent experiments with ±SD. c A comparison and time-course of Olaparib (10 μM) induced TBK-1 and IRF-3 phosphorylation between control and GRB2-KO HeLa cells. d GRB2 re-expression in GRB2-KO HeLa cells suppressed Olaparib (10 μM, 48 h) induced TBK-1 and IRF-3 phosphorylation. e Representative immunofluorescence images showing olaparib (10 μM, 48 h) treatment induces nuclear accumulation of phosphorylated IRF-3 (pIRF-3) in GRB2-KO HeLa cells. Scale bar 10 μm. f The increased TBK-1 and IRF-3 phosphorylation observed in GRB2-KO HeLa cells are independent of RAS (PD184352; left panels) or Akt (MK2206; right panels) signaling. g The increased TBK-1 and IRF-3 phosphorylation in GRB2-KO HeLa cells are MRE11 dependent, n = 3. h STING knockdown is sufficient to abrogate Olaparib (10 μM) induced TBK-1 and IRF-3 phosphorylation in GRB2-KO HeLa cells. n = 3. i The increased TBK-1 and IRF-3 phosphorylation in GRB2-KO HeLa cells are STING dependent, n = 3. ik qRT-PCR showing Olaparib induce increased level of INF-B, CCL5 and CXCL-10 mRNA in HeLa cells with ±SD, n = 3. l Quantitation of cytokines array results from two independent experiments showing upregulated inflammatory cytokine released in culture medium with olaparib treatment, n = 2. m Recruitment of cytotoxic CD8 + T-lymphocytes (CTL) isolated from PBMC to GRB2-KO HeLa cells in response to Olaparib treatment, n = 3, ±SD are shown. n siRNA mediated STING knockdown in GRB2-KO HeLa cells abrogate PBMC isolated CD8 + CTL recruitment, n = 3, errors are in ±SD. The significance was analyzed by two-sided Student’s t test. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, and ****P ≤ 0.0001; NS not significant. For (bn), source data are provided as a Source Data file.

Production of excessive micronuclei in PARPi-treated GRB2-KO cells activated the cGAS/STING pathway, as measured by increased phosphorylated TBK1 (p-TBK1) and its downstream target IRF3 phosphorylation (pIRF3) levels. Both p-TBK1 and pIRF3 were measurably upregulated in PARPi treated GRB2-KO cells in all tested time periods (Fig. 4c and Supplementary Fig. 7c, d). Importantly, reconstitution of GRB2 in GRB2-KO cells restored p-TBK1 and p-IRF3 level comparable to the control (Fig. 4d and Supplementary Fig. 7e, f). These data imply GRB2 expression alone is sufficient to restrict cytoplasmic DNA accumulation.

Under normal conditions IRF-3 resides in the cytoplasm of cells but phosphorylation triggered by cytoplasmic DNA leads to its translocation to the nucleus. This phosphorylation and translocation in association with the p300/CBP coactivator protein promotes DNA binding and transcriptional activity47. We therefore sought to determine if the functionally important p-IRF3 nuclear translocation occurred. Immunofluorescence analysis revealed a drastic increase in pIRF3 level in the nucleus of GRB2-KO cells treated with PARPi (Fig. 4e). These data are consistent with western blot analysis and further imply increased transcriptional activity of IRF3 in GRB2-KO cells in response to PARPi treatment.

As GRB2 acts in the RAS and PI3-Kinase signaling pathway48, we tested to see if the observed cGAS/STING upregulation is due to its effect on cytoplasmic signaling. Control and GRB2-KO cells were treated with MEK and AKT inhibitors and the pTBK1 and pIRF3 levels were measured. The results showed that inhibition of RAS-MAPK or PI3-K signaling pathways have little effect on cGAS/STING signaling (Fig. 4f and Supplementary Fig. 7g, h). Thus, the pTBK1 response observed here corresponds to GRB2 function in DNA replication stress.

In GRB2-depleted cells, MRE11 resected stalled replication forks (Fig. 1g, h). We therefore tested if the observed pTBK1 and pIRF3 upregulation in GRB2-KO cells correlates with MRE11 expression level. We knocked down MRE11 (siMRE11) in GRB2-KO cells and measured TBK1 and IRF3 phosphorylation. The depletion of MRE11 in GRB2-KO cells inhibited PARPi induced pTBK and pIRF3 phosphorylation (Fig. 4g and Supplementary Fig. 7i). These results suggest that the elevated TBK1 and TRF3 phosphorylation in GRB2-KO cells is caused by cytoplasmic DNA generated by MRE11 nucleolytic processing in response to PARPi-induced replication stress. To test if inhibiting MRE11 nuclease activity can also inhibit PARPi-induced pTBK1 in GRB2-KO cells, we treated control and GRB2-KO cells with MRE11 inhibitors Mirin with and without olaparib. Our results showed MRE11 inhibitors did not fully recapitulate the MRE11-KD phenotype in GRB2-KO cells (Supplementary Fig. 7j–l). Although there was a measurable pTBK1 reduction in the combined MRE11i and PARPi treated cells, it was not as effective as MRE11-KD. Thus, MRE11i may either lack sufficient potency to provide full protection against degradation of an unprotected replication fork without GRB2 being present or MRE11i promotes alternative activities at forks compared to MRE11-KD in the absence of GRB2.

TBK1 and IRF3 phosphorylation can be upregulated by STING or MAVS49,50. To elucidate which pathway activation resulted in TBK1 and IRF3 phosphorylation, we used shRNA knock down of either STING or MAVS to show that the cGAS/STING, but not the MAVS, signaling pathway is activated in GRB2-KO cells treated with PARPi (Fig. 4h and Supplementary Fig. 8a–c).

We next tested and found cGAS/STING activation correlated with upregulation of type I IFNs and pro-inflammatory cytokine mRNA. In GRB2-KO cells, IFNβ, CCL2 and CXCL10 mRNA were upregulated in response to PARPi treatment (Fig. 4i, k). To determine if the observed mRNA upregulation correlated with secretion of these chemoattractants, we used a human inflammation antibody array to measure cytokines into the extracellular environment. In response to PARPi treatment, GRB2-KO cells secreted a higher concentration of chemoattractant cytokines for T-cells (CCL5), myeloid and dendritic cells (CCL2), T-cells and NK-cells (CXCL 10) (Fig. 4l and Supplementary Fig. 8d). We also observed a significant increase in the secretion of PDGF-BB, a known mitogen and a chemoattractant51.

We therefore investigated if the released cytokines from GRB2-KO cells are sufficient to attract cytotoxic CD8+ cytotoxic T-cells. Peripheral blood mononuclear cells (PBMC) were isolated and tested in a cell-migration recruitment assay. Only GRB2-KO cells that were treated with PARPi showed a significant increase in cytotoxic T-cell infiltration compared to untreated and the control cells with normal level of GRB2 (Fig. 4m). Knockdown of STING in GRB2-KO cells abrogated CD8+ cytotoxic T-cells infiltration (Fig. 4n).

These collective and complementary data support a key role of nuclear GRB2 in the DNA RF stress response. In cultured HeLa and HAP-1 cells under replication stress induced by PARPi, GRB2 deficiency causes replication fork defects leading to genomic instability and cytosolic DNA accumulation. This then activates cGAS/STING leading to production of chemoattractant cytokines, which in turn recruit cytotoxic T-cells.

PARPi averts tumor progression in a GRB2-depleted ovarian cancer model

Having established functional and mechanistic roles for GRB2 during RF stress that restrict inflammatory cytokine release, we directly investigated immune surveillance of PARPi-treated cancer cells with low GRB2 in a whole animal setting. Our prior cancer database analyses suggested that ovarian cancer with low GRB2 expression correlated with increased cytotoxic T-cell infiltration52. We therefore tested this notion experimentally by using metastatic ovarian cancer cell-line ID8. We generated Grb2-KD ID8 mouse cells and found that PARPi also induced TBK1 phosphorylation (Supplementary Fig. 8e–g). In preparation for oral dosing, we also compared TBK1 phosphorylation level between Olaparib and Talazoparib, where Talazoparib induced higher TBK1 response with lower dose (Supplementary Fig. 8f, g) establishing it as a suitable substitution for Olaparib in mice. Grb2-KD mice receiving Talazoparib showed a reduced cancer burden (Fig. 5a and Supplementary Fig. 9a). This was recapitulated in our tumor volume measurements based on total flux (photons per second) measurements. Grb2-KD cells treated with Talazoparib also showed a significantly slower tumor growth rate (Fig. 5b). Furthermore, Grb2-KD mice treated with Talazoparib survived significantly longer than those treated with PBS or the control cells irrespective of treatment conditions (Fig. 5c).

Fig. 5: GRB2 depletion sensitizes cells to PARPi.
figure 5

a A schematic experimental overview for monitoring cancer growth with luciferase imaging. Below, Representative images of luciferase expression ID8 cells detected on day-7 and day-28. Talazoparib (0.33 mg/kg) treated control and GRB2-KD mice are only shown. b Quantitative luciferase signal detection using IVIS imaging for n = 5 mice per group up to 28 days, the mean with ±SD are shown. c Kaplan–Myer curve showing survival of mice in the four-treatment group. d The measurements of Ascites volume n = 4 mice all 4 treatment groups, the mean with ±SD are shown. e, f Measurement of mouse IL-2 level and IL-12 in the Ascites fluids of n = 4 mice, the mean with ±SD are shown. The significance was analyzed by two-sided Student’s t test. *P ≤ 0.05, **P ≤ 0.01 and ***P ≤ 0.001; NS not significant. For (bf), source data are provided as a Source Data file.

A major complication associated with metastatic ovarian cancer is ascites buildup, which is often associated with advanced disease53. Grb2-KD cells injected mice treated with PARPi showed significantly lower body weight due to reduced ascites volume compared to the control mice and to vehicle treated groups (Fig. 5d, and Supplementary Fig. 9b, c). The inflammatory cytokine interleukin 12 (IL-12) is often associated with cytotoxic T-cell and NK cell activation54, so we measured the IL-12 and IL-12 p40 level in the ascites fluid. We found significant elevation in mice bearing Grb2-KD cells and treated with PARPi indicative of high immune cell activities (Fig. 5e, f). These collective data show that low GRB2 levels enable cGAS/STING activation and immune detection of cancer cells.