Abstract Liver metastasis (LM) confers poor survival and therapy resistance across cancer types, but the mechanisms of liver-metastatic organotropism remain unknown. Through in vivo CRISPR-Cas9 screens, we found that Pip4k2c loss conferred LM, but had no impact on lung metastasis or primary tumor growth. Pip4k2c-deficient cells were hypersensitized to insulin-mediated PI3K/AKT signaling and exploited the insulin-rich liver milieu for organ-specific metastasis. We observed concordant changes in PIP4K2C expression and distinct metabolic changes in 3,511 patient melanomas, including primary tumors, LMs and lung metastases. We found that systemic PI3K inhibition exacerbated LM burden in mice injected with Pip4k2c-deficient cancer cells through host-mediated increase in hepatic insulin levels, however, this circuit could be broken by concurrent administration of an SGLT2 inhibitor or feeding of a ketogenic diet. Thus, this work demonstrates a rare example of metastatic organotropism through co-optation of physiological metabolic cues, and proposes therapeutic avenues to counteract these mechanisms. Introduction Metastasis is a major determinant of mortality in patients with cancer^[112]1, and metastatic patterns have important clinical and therapeutic implications^[113]2. Liver metastasis (LM) occurs frequently across different cancer types, including melanoma, and carcinomas of the colon, pancreas and breast^[114]3 and confers a poor prognosis and reduced response rates to modern cancer therapies, such as immune checkpoint blockade (ICB), when compared to response rates in other common metastatic sites, such as the lung^[115]3–[116]5. The underlying mechanisms for these clinical phenotypes are poorly understood. This is in part due to difficulties in modeling of salient molecular underpinnings of metastatic organotropism pre-clinically. Despite extensive efforts in a variety of disease contexts, divergent somatic mutations that fully explain organ-specific metastasis have not yet been identified^[117]6,[118]7. While this may be due to the limited power of individual studies and extensive disease heterogeneity, it is likely that other biological factors underly cancer organotropism. Indeed, recent work in melanoma patients, for example, revealed a higher rate of chromosomal instability, a neural-like cell state and metabolic adaptation enriched in the brain metastases compared to extra-cranial metastases^[119]8. Whether similar organ-specific mechanisms exist for other sites is unknown. The liver is the major metabolic organ, where a multitude of cues are generated, received, and integrated, to regulate the supply of systemic nutrients, such as glucose and fatty acids^[120]9. Additionally, the liver has a dual blood supply via the hepatic artery and the portal vein, each of which may be a route for metastatic spread. There is also increasing evidence for hepatic modulation of local and peripheral immunity^[121]10,[122]11. These and other distinct organ features may contribute to the liver-metastatic niche in melanoma, and other cancers. To identify determinants of liver-metastatic organotropism in an unbiased fashion, we leveraged a syngeneic murine melanoma model, and performed large-scale CRISPR-Cas9 screens. We identified loss of Pip4k2c as driver of liver- but not extra-hepatic metastasis. We showed that this organotropism is due to co-optation of the insulin-rich liver milieu, to which Pip4k2-deficient cells are hypersensitized to, and unraveled downstream signaling, and metabolic and transcriptomic adaptations enriched in liver metastases in mouse and patient tumors. We observed a paradoxical increase in LM burden in animals treated with systemic PI3K inhibitors and showed that this was due to host-mediated responses that exacerbated the insulin-gradient, thereby promoting metastatic selectivity of Pip4k2c-deficient cells to the liver. Combination of PI3K inhibition with either systemic SGLT2 inhibition or feeding animals a ketogenic diet, reduced LM burden, while having little effect on other metastatic sites or primary tumor growth. Analyses and additional functional validation in prostate cancer and colorectal cancer models indicate a similar role of Pip42kc in conferring liver metastasis, suggesting that the proposed biological and therapeutic principles may extend beyond melanoma. Results CRISPR screens reveal determinants of liver metastasis To identify drivers of liver metastasis, we used a syngeneic melanoma model, HCmel12, which was derived from Hgf/Cdk4^R24C mice that spontaneously develop melanoma^[123]12 and recapitulates the potential for liver metastasis and therapeutic response patterns seen in patients with metastatic melanoma^[124]13,[125]14. We performed a large-scale in vivo CRISPR-Cas9 screen in Cas9-expressing HCmel12 cells, perturbing 713 kinases in the mouse genome with 2,852 single-guide RNAs (sgRNAs) (four per target gene, and 100 non-targeting control guides using the “Brie” library^[126]15) ([127]Fig. 1a, [128]Extended Data Fig. 1a–[129]c). The edited cell pool (Cas9 + Brie), as well as Cas9-expressing cells without sgRNAs and parental cells, were injected via tail vein or subcutaneously implanted in C57BL/6 (B6) animals. In parallel, edited cell lines were maintained in vitro and collected at several time points for the duration of the experiment ([130]Methods). Dissection of animals ([131]Methods) revealed that the Cas9 + Brie group had a significantly higher rate of animals with liver metastasis ([132]Fig. 1b–[133]d), higher metastasis count per animal ([134]Fig. 1e), and increased liver metastatic burden compared to the Cas9 and parental groups ([135]Fig. 1f), while there was no difference in lung metastatic burden across these groups ([136]Fig. 1g). This suggested that loss of kinases targeted by the Brie sgRNA library enhanced the liver metastatic potential of HCmel12 melanoma cells. Figure 1. In vivo CRISPR-Cas9 screen identifies drivers of liver metastasis. Figure 1. [137]Open in a new tab a, Experimental design of in vivo CRISPR-Cas9 screen. b, Photographs of representative livers removed from animals in different groups as indicated. Yellow arrowheads indicate pigmented and non-pigmented liver metastases. c, Hematoxylin and Eosin staining of representative sections of livers from indicated groups, scale bar 2mm, 40x magnification (n=2 independent experiments). d, Stack bar plots indicate fraction of animals bearing liver metastases across different experimental groups. e,f Liver metastasis count (e) and calculated liver metastasis disease burden (f) across experimental groups. g, Lung metastasis count in corresponding animals from (f); (d-g), n=9 mice per group; bars represent mean ± s.e.m.; One-Way ANOVA Tukey’s multiple comparisons test. Data is representative of two independent experiments (d-g). Significance levels as indicated on top of each comparison. h, Exemplary distribution of sgRNA read fractions in three animals from the in vivo CRISPR-Cas9 metastasis screen. Pie charts show the fraction of the most abundant sgRNAs (sgRNAs with >2% of total reads) in each individual liver metastatic lesion next to each animal. i, Summary of fractions of enriched sgRNAs across all lesions from (h) within the individual mouse (larger pie charts below each animal). Before identifying perturbations that may alter liver metastatic potential, we first assessed the sgRNA library complexity within the plasmid pool, edited cells, transplanted primary tumors, and distant metastasis (liver, lung and lymph nodes). Compared to the plasmid pool, cell line input, and primary tumors, which showed preserved and co-correlated library diversity, sgRNA diversity decreased substantially in metastatic lesions ([138]Extended Data Fig. 1d–[139]e) across different sites, while samples from the same metastatic sites showed stronger co-correlation ([140]Extended Data Fig. 1d). In line with this, principal component analysis (PCA) of sgRNA representations revealed clades of metastases and transplanted tumors/cell lines along the PC1 (explaining 49.7% of variability) and PC2 (5.4 % of variability) ([141]Extended Data Fig. 1f). Next, we identified sgRNAs targeting essential genes (e.g., Plk1, Cdk7 and Cdk12) that were depleted after 14 days of editing in cells prior to injection into animals ([142]Extended Data Fig. 2a–[143]c, [144]Supplementary Table 1). Additionally, sgRNAs targeting Cdk4, a known dependency in HCmel12, were also significantly depleted (log2FC=-10.206, FDR<0.06). Compared to edited cells prior to injection, primary tumors showed significant depletion of multiple genes, including Jak1 and Jak2, suggesting that the Jak/Stat pathway may be important for tumor growth, as absence of intact Jak1/Jak2 has been implicated in immune evasion^[145]16. Stk11 was also depleted in primary tumors; loss of Stk11 (encoding for Lkb1) promotes metastasis and confers therapy resistance^[146]17. The most strongly enriched perturbation involved in primary tumor growth was loss of Dapk3 (log2FC=4.3693, FDR<0.007) ([147]Extended Data Fig. 2b, [148]Supplementary Table 2), a putative tumor suppressor gene whose deleterious mutations or loss occurs in up to ~7% of melanomas^[149]18. Together, these data suggested that the conducted screen was robust and yielded known and unknown biology, with sgRNA diversity reducing along the metastatic progression axis. This observation suggested that a small fraction of perturbations may determine metastatic potential^[150]19. We next sought to identify sgRNAs associated with metastatic potential to the liver. We used three criteria to nominate such putative drivers: sgRNAs that (1) are enriched with an FDR <0.1 and/or (2) account for >2% of sequencing reads within a given lesion and/or (3) are enriched in two or more biological replicates. We identified 80 sgRNAs that meet one (65), two (17) or all (2) of these criteria ([151]Supplementary Table 3). Among the genes targeted by these sgRNAs are those involved in cell motility (Tesk1, Pkn1, Mylk4)^[152]20–[153]22, nucleotide homeostasis and metabolism (Prps1, Prps2, Adck2, Nme8)^[154]23, modulation of insulin sensing (Trib1, Pip4k2c)^[155]24–[156]26, or function as tumor suppressors (Fgfr3)^[157]27, such that the loss of these genes would promote hallmarks of metastasis. The “top hit” in this screen by all applied metrics was loss of Pip4k2c which encodes Phosphatidylinositol-5-Phosphate 4-Kinase Type 2 Gamma. Specifically, loss of Pip4k2c was the most enriched perturbation in five of 14 (~36%) investigated liver metastatic lesions. In all of these lesions, sgRNAs targeting Pip4k2c accounted for the majority of sequencing reads, while in four of these five lesions, this perturbation accounted for >98% of sequencing reads ([158]Fig. 1h,[159]i, [160]Extended Data Fig. 2d). Furthermore, loss of Pip4k2c was the only perturbation (at an FDR <0.06), that was highly specific to liver metastasis ([161]Extended Data Fig. 2e). These findings suggest that Pip4k2c loss may uniquely promote liver metastasis. Pip4k2c loss sensitizes cells to insulin-mediated signaling Pip4k2c is part of the Pip4k family that consists of three isoforms in both mice and humans^[162]24. The role of these kinases is poorly understood, but recent work suggests that Pip4k’s, and the gene product of Pip4k2c, suppress Pip5k activity in response to insulin. This results in reduced production of phosphatidylinositol-4–5-bisphosphate (PI (4,5) P[2]), the substrate of PI3 Kinase (PI3K), which converts PI (4,5) P[2] to phosphatidylinositol-3-4-5-triphosphate (PI (3,4,5) P[3]) to subsequently activate AKT. Thus, we hypothesized that loss of Pip4k2c would lead to hyperactivation of the PI3K/AKT pathway in response to insulin. We hypothesized that because the liver is the organ with the highest insulin concentration (outside of the pancreas, where it is produced), Pip4k2c-loss mediated PI3K/AKT hypersensitization to insulin in tumor cells could encourage formation of an organ-specific metastatic niche. To test whether Pip4k2c promotes PI3K/AKT hypersensitivity to insulin in melanoma cells, we generated HCmel12-Pip4k2c-KO cell lines (Pip4k2c^KO) using four different sgRNAs, including the top enriched sgRNA in our in vivo screen ([163]Extended Data Fig. 3a–[164]b). We next determined the impact of Pip4k2c^KO on downstream PI3K/AKT pathway activation in response to insulin. Compared to parental cells, insulin stimulation of Pip4k2c^KO resulted in higher and prolonged induction of phosphorylated AKT in both murine HCmel12 and the human melanoma model A375 ([165]Fig. 2a,[166]b, [167]Extended Data Fig. 3c–[168]e), increased phosphorylation of insulin receptor (INSR) ([169]Extended Data Fig. 3f) and showed increased cell migration capacity, while the proliferation rate was not altered ([170]Extended Data Fig. 3g–[171]j). Bulk RNA-sequencing of parental compared to Pip4k2c^KO cells stimulated with insulin revealed enrichment of mTOR pathway activity ([172]Extended Date Fig. 3k, [173]Supplementary Table 4). We rescued Pip4k2c^KO with one of two open reading frames (ORFs): full-length (Pip4k2c^Rec) or allosteric-domain deficient (aa69–75 (VMLLPDD → EIFLPNN)) (Pip4k2c^AD), which was previously implicated in Pip4k2c function^[174]24 ([175]Extended Data Fig. 3l–[176]n). While Pip4k2c^AD continued to demonstrate high baseline and insulin-mediated Akt phosphorylation, Pip4k2c^Rec had lower p-Akt levels in both human and murine melanoma models ([177]Extended Data Fig. 3l–[178]n). This suggests that lack of the allosteric domain, but not the kinase domain of Pip4k2c, is sufficient to confer hypersensitization to insulin; this finding is consistent with prior work showing that Pip4k2c has very poor kinase activity^[179]24. Treatment with pan-PI3K inhibitor GDC-0941 (pictilisib) (with and without insulin stimulation) ([180]Extended Data Fig. 3l–[181]n) completely abrogated baseline Akt phosphorylation in cells with all of these genotypes, while insulin partly rescued PI3K activity ([182]Extended Data Fig. 3l–[183]n), which is consistent with a prior report^[184]28. We also tested PI3K alpha inhibitor BYL-719, which overall showed lower efficacy but similar results for insulin-mediated bypass activation ([185]Extended Data Fig. 3l–[186]n). Figure 2. Loss of Pip4k2c enhances sensitivity to insulin and promotes liver metastasis, but not lung metastasis. Figure 2. [187]Open in a new tab a,b, Immunoblots showing phosphorylated and total protein of key signaling nodes of pAkt^S473 and pAkt^T308 in WT or Pip4k2c^KO cells in (a) HCmel12 and (b) A375 melanoma cell models with (+) or without (-) insulin (250ng/ml) stimulation. c,d, Liver metastasis disease burden (in mm^3) and lung metastases count following tail vein injection of parental and Pip4k2c KO HCmel12 melanoma in C57BL/6 mice. e,f, Liver and lung metastasis burden following tail vein injection of parental and PIP4K2C KO A375 melanoma cells in NSG mice. g, Incidence of macroscopic liver metastasis in animals following orthotopic injection of parental or Pip4k2c KO CT26 colorectal cancer cells in BALB/c mice. h, Comparison of tumor weight of primary tumors from experiment shown in (g); (c-f) n=10 mice per group; (g-h) n=4–5 mice per group; mean ±s.e.m. One-Way ANOVA Tukey’s multiple comparisons test for (c,d) and Mann-Whitney Two-tailed test for (e-g). Data are representative of three (c,d), two (e,f) or one (h,g) independent experiments. Significance levels as indicated on top of each comparison. Loss of Pip4k2c confers liver-metastatic organotropism To validate the role of Pip4k2c loss in vivo, we next injected either parental (Pip4k2c^WT), Pip4k2c^KO, Pip4k2c^Rec, or Pip4k2c^AD HCmel12 melanoma cells via tail vein in B6 mice. Pip4k2c^KO resulted in a significantly increased LM burden that was rescued by Pip4k2c^Rec, but not Pip4k2c^AD ([188]Fig. 2c). Importantly, none of these perturbations altered lung metastatic burden ([189]Fig. 2d). Consistently, loss of PIP4K2C in human melanoma cell line A375 conferred significantly increased metastasis to the liver, but not to the lung ([190]Fig. 2e,[191]f, [192]Extended Data Fig. 3o). To determine whether Pip4k2c loss plays a role in LM development in other cancers that also frequently metastasize to the liver, we generated Pip4k2c KO in a colorectal cancer cell line (CT26) and orthotopically implanted these or parental cells into the colonic submucosa of BALB/c mice. Primary tumors arising from parental CT26 resulted in LM in 20% while Pip4k2c^KO cells gave rise to LM in 75% of animals ([193]Fig. 2g). Notably, Pip4k2c^KO gave rise to smaller primary colorectal tumors ([194]Fig. 2h), thus, indicating that higher rates of LM development were due to increased invasive capacity, as observed in melanoma models ([195]Fig. 2c,[196]e). Together, these results suggest that loss of Pip4k2c hypersensitizes cancer cells to insulin mediated PI3K/AKT hyperactivation, and thereby mediates metastasis specifically to the liver, but not to the lung. Systemic PI3K inhibition enhances LM through host effects. Since PI3K-i could abrogate PI3K/AKT signaling in vitro ([197]Extended Data Fig. 3m–[198]n), we reasoned that systemic treatment with the same drug would reduce LM burden in vivo. We injected parental or Pip4k2c^KO cells and treated animals with either vehicle control or GDC-0941 ([199]Methods). Surprisingly, we found that liver-metastatic burden was significantly increased in animals bearing Pip4k2c^KO tumor cells, while lung metastatic burden was not altered ([200]Fig. 3a,[201]b). We reasoned that this paradoxical increase in LM burden in Pip4k2c^KO tumor bearing mice in response to PI3K inhibition may be explained by the effects of the drug on the host, which may promote liver metastasis in an insulin-dependent manner. Indeed, treatment with GDC-0941 resulted in a significant increase in blood glucose levels that peaked at 60 minutes after administration and was sustained for at least 90 minutes ([202]Fig. 3c). Accordingly, the levels of C-peptide, a short amino-acid polypeptide of pro-insulin and equimolar surrogate for insulin production, were significantly increased in GDC-0941 treated animals with either Pip4k2c^WT or Pip4k2c^KO bearing tumors ([203]Fig. 3d, [204]Extended Data Fig. 3p), suggesting that these differences in metastatic burden are explained by host responses to this treatment. To assess whether GDC-0941 induced glucose and insulin spikes influenced metastatic tumor cell sensitivity to insulin in situ, we treated Pip4k2c^KO liver-metastasis bearing mice with GDC-0941 or vehicle control. After 60 minutes when blood glucose concentration peaks following PI3K-i treatment, we performed [F18] fluorodeoxyglucose micro positron-emission tomography (FDG μPET). We found that Pip4k2c^KO liver metastases had a 3-fold increase in FDG uptake in PI3K-i treated mice ([205]Fig. 3e, [206]f). Lastly, we directly measured homeostatic levels of insulin in livers and lungs, confirming significantly higher levels in the hepatic environment ([207]Fig. 3g). If high insulin concentration in the liver is an important cue for liver-metastatic organotropism, we reasoned that increasing insulin levels in the lung should increase the rate of metastasis to this organ. To test this, we subcutaneously implanted mice with slow-release insulin pads ([208]Fig. 3h), which continuously deliver high systemic levels of insulin, followed by injection of cancer cells via tail vein, and determined liver and lung metastatic burden. While insulin delivery resulted increased metastatic burden was increased in both liver and lung ([209]Fig. 3i), there was a significantly elevated lung-metastatic potential compared to control pad-implanted animals ([210]Fig. 3j, [211]k), suggesting that some disease burden was diverted to the lungs with higher local insulin levels. Together, these results suggest that Pip4k2c^KO cells efficiently co-opt the insulin-rich liver milieu for metastasis, and this can be further enhanced by host responses to PI3K inhibition. Figure 3. Systemic feedback loop induced by PI3K inhibitor treatment in vivo promotes liver organotropism in Pip4k2c-deficient tumors in an insulin-dependent manner. Figure 3. [212]Open in a new tab a, Liver metastasis disease burden (in mm^3) upon tail vein injection of parental and Pip4k2c KO HCmel12 melanoma cells with and without GDC-0941 treatment (daily, 5 days a week, 100mg/kg). b, Lung metastasis burden (metastasis count) in corresponding animals from (a). n = 10 mice per group; mean ±s.e.m; c, Blood glucose levels (in mg/dL) in mice following treatment with GDC-0941 or vehicle control over time (minutes). D, C-peptide levels (in pM) in corresponding animals from (c); n = 10 mice per group; mean ±s.e.m; 2-way ANOVA Tukey’s multiple comparisons test for (a-b) and Mann-Whitney Two-tailed test for (c-d). Significance levels as indicated. E, f, Quantification of FDG-μPET (18F-Fluorodeoxyglucose in per cent of injected dose (ID) per gram of (e)) and corresponding liver metastasis uptake (f) 60 min after GDC-0941 administration or vehicle control. Yellow arrows indicate individual liver metastatic lesions. Cardiac FDG-uptake is visualized (asterisk); n=3 mice per group, mean ±s.e.m; unpaired two-sided-t-test (e). g, Insulin levels (in pg/ul) in liver and lung tissues from non-tumor bearing mice. n=10 mice per group, mean ±s.e.m; Mann-Whitney Two-tailed test. h, Schematic illustrating the insulin pad experiment in mice; i, j, Percentage of metastasis surface area in liver (i) and lung (j) of animals implanted with a control pad or insulin pad, and followed by injection of tumor cells via tail vein in NSG mice. k, Ratio of lung over liver metastatic burden from experiment in (i,j); n=5 mice per group; mean ± s.e.m; Mann-Whitney Two-tailed test; Data are representative of three (a-d), two (e-k) independent experiments. Significance levels as indicated on top of each comparison. Human and murine LM exhibit distinct metabolic adaptations To further examine cancer cell states in liver compared to lung metastases, we performed single-cell RNA-sequencing (scRNA-seq) of eight mice with concurrent liver and lung metastases. Following rigorous quality control ([213]Methods, [214]Supplementary Tables 4 and [215]5), a total of 95,606 cell transcriptomes were included for downstream analyses. We identified tumor cells by their expression of melanocytic lineage markers, including Mitf, Mlana, Dct, Tyr, and Tyrp1. Differential gene expression and subsequent GSEA of melanoma cells revealed strong enrichment of metabolic pathways in cells isolated from liver metastases, including regulation of oxidative phosphorylation ([216]Fig. 4a, [217]Extended Data Fig. 4a–[218]g). In support of the observations in melanoma, we analyzed another bulk RNA-seq data set of a prostate cancer model^[219]29 that develops metastasis to multiple organs, including to liver, lung, and bone, and find the same metabolic pathway enrichment in liver compared to lung metastases ([220]Fig. 4b). Figure 4. multi-omics analysis of human and mouse liver and lung metastasis. Figure 4. [221]Open in a new tab a, Top enriched pathways (adjusted p value < 0.05, top 5 pathways per gene set ranked by NES) of cancer cells isolated from concurrent liver (n=8 specimens, pink) vs lung (n=8 specimens, purple) metastases in mice injected with HCmel12 melanoma cells. b, Pathway enrichment (FDR q-value < 0.05) within Arriaga et al., cohort comparing liver (n=7 specimens, pink) vs. lung (n=14 specimens, purple) metastases in a prostate cancer metastasis model. c, Enriched pathways (FDR q-value < 0.05) in liver (n=78 patients, Met500; n=15 patients, LM15, pink) vs lung (n=16 patients, Met500, purple) metastases across tumors. d, Enriched pathways (adjusted p value < 0.05) in liver (n=364) vs lung (n=743) metastases in patients with melanoma tumors; FDR value for GSEA was obtained by Benjamin-Hochberg correction. e,f, Expression of PIP4K2C in melanoma patient tumors comparing primary cutaneous tumors (n=2,404 patients) vs liver (n=364) (e), and liver (n=364 patients) vs lung (n= 743 patients). Mann-Whitney Two-tailed test. g, Expression of Pip4k2c from the Arriaga et al., cohort comparing liver (n=7 specimens) vs lung (n=14 specimens), Wilcoxon rank-sum test. For all boxplots, n refers to the number of samples, and p refers to the p-value. The center line indicates the median, the box limits denote the first and third quartiles, and the whiskers indicate the lowest or highest data points at the first quartile minus or plus 1.5 times the interquartile range; Significance levels as indicated. h, Cumulative results of selected top differentially abundant metabolites in animals with liver and/or lung metastases following injection with either parental (PIP4K2C^WT) or PIP4K2C^KO A375 human melanoma cells; n=5 mice per group; unpaired two-sided-t-test. Lactate (p=2.36727E-05), Pyruvate (p=0.03), Citrate (p=0.25), a-Ketoglutarate (p=0.18), Succinate (p=0.004), Fumarate (p=0.04), Malate (p=0.04), Hexose (p= 6.72275E-06). To determine the relevance of these phenotypic differences in a patient context, we performed RNA-sequencing of melanoma LMs and combined these with publicly available data ([222]Methods)^[223]30, totaling 252 metastasis transcriptomes, including 93 liver metastases ([224]Supplementary Tables 4 and [225]5). Comparison of LM transcriptomes with lung metastases revealed strong enrichment of metabolic pathways in LM, including TCA cycle, OxPhos, and mTORC1 signaling ([226]Fig. 4c, [227]Extended Data Fig. 4 h–[228]j, and [229]Supplementary Tables 4 and [230]5). These results were maintained when including only samples with high purity, reducing potential transcriptome contamination by healthy hepatocytes ([231]Extended Data Fig. 4j,[232]k, [233]Supplementary Tables 4 and [234]5). Additionally, our scRNA-seq pathway analyses were exclusively performed on tumor cells yielded comparable results. To comprehensively determine the role of these observations in a large real-world data set, we analyzed 3,511 cutaneous melanoma patients, including 2404 primary cutaneous melanomas (PCMs), 364 LMs and 743 lung metastases ([235]Supplementary Table 4). For all specimens, tumor regions were enriched through macro-dissecting and profiled using RNA-sequencing and exome sequencing (either whole-exome or 592 gene panel DNA-sequencing) at Caris Life Sciences (Phoenix, AZ). We found strong enrichment of metabolic pathways, including TCA cycle and OxPhos in LMs compared to PCMs and lung metastases ([236]Fig. 4d), which mirrored our observations in mouse models ([237]Fig. 4a,[238]b) and publicly available patient data analyzed here ([239]Fig. 4c). Overall, these transcriptomics data suggest that cancer cells in the liver enrich for key metabolic pathways when compared to other metastatic sites, both in melanoma and other cancers. In the same patient cohort, we found that PIP4K2C, but PIP4K2A or PIP4K2B, had a lower expression in LMs compared to PCMs and lung metastases (p<0.0001 and p=0.08, respectively) ([240]Fig. 4e, [241]f. [242]Extended Data Fig. 4l. [243]m). Additionally, comparison of genomics of LMs with either PCM or lung metastases in these patients showed distinct distribution of common oncogenic driver mutations, including BRAF, NRAS and NF1, and revealed an enrichment of the less commonly mutated (in melanoma) gene PIK3CA encoding for p110α, the catalytic subunit of PI3-kinase in LMs compared to PCMs and lung metastases (4.27% vs. 1.21%, p=0.003, and 4.27% vs. 2.34%, p=0.19, respectively) ([244]Extended Data Figure 4n). Similarly, we found lower expression of Pip4k2c, but not Pip4k2a/b, in LMs compared to other metastatic sites in the Arriaga et al., prostate cancer metastatic mouse model data ([245]Fig. 4g, [246]Extended Data Fig. 4o). We next sought to directly measure potential differences in metabolites between liver and lung metastases. For this purpose, we injected A375 cells (parental or Pip4k2c^KO) and collected liver and lung metastases ([247]Methods). We performed mass spectrometry on five liver and five concurrent lung metastases from animals injected with Pip4k2c^KO cells, and three liver and five lung metastases from mice injected with Pip4k2c^WT cells. Principal component analysis (PCA) demonstrated that across and within each genotype, disease site (liver vs. lung) was the most important driver of variability in metabolic profiles ([248]Extended Data Fig. 5a,[249]b, [250]Supplementary Table 6). Several key metabolites of the TCA cycle, including a-ketoglutarate, succinate, fumarate and malate were found at higher abundances in liver compared to lung metastases ([251]Figure 4h, [252]Extended Data Fig. 5c–[253]e), consistent with the findings indicated from our transcriptomics analyses ([254]Figure 4a–[255]d). INSR is necessary for liver-metastatic organotropism Our data suggest that co-optation and metabolic adaptions to the insulin-rich liver milieu are strongly associated with liver-metastatic organotropism. To determine whether insulin acting on cancer cells is necessary for both liver-metastatic organotropism and increased disease burden in response to systemic PI3K inhibition, we transduced parental or Pip4k2c^KO cells with a doxycycline(dox)-inducible short hairpin RNA (shRNA) targeting the insulin receptor (Insr) to generate otherwise isogenic, conditional Insr knockdown (Insr^shIR) or Pip4k2c^KO/Insr^shIR cells ([256]Fig. 5a). As expected, we found a significantly impaired induction of Akt phosphorylation in response to insulin in both dox-induced Insr^KD and Pip4k2c^KO/Insr^shIR cells, compared to their non-dox treated counterparts. Across all conditions, GDC-0941 effectively abrogated p-AKT ([257]Extended Data Fig. 6a). Next, we injected mice with dox-treated or untreated Pip4k2c^KO/Insr^shIR tumor cells followed by treatment with vehicle or GDC-0941. Insr knockdown alone resulted in LM reduction compared to non-Dox treated animals (albeit not statistically significant), and in mice treated with GDC-0941, Insr knockdown in Pip4k2c^KO tumor cells resulted in a significantly reduced LM burden ([258]Fig. 5b). Across all conditions, there was no change in lung metastasis burden ([259]Fig 5c). Overall, these results suggest that while GDC-0941 effectively abrogates Akt phosphorylation in vitro, host responses to the drug and subsequent insulin-dependent circuitry increase metastasis to the liver, but not in the lung. Figure 5. Genetic, dietary or pharmacological strategies disrupting systemic host effects of PI3K-inhibition reduces metastatic liver organotropism. Figure 5. [260]Open in a new tab a, Immunoblot probing Insr in Pip4k2c^KO/Insr^shIR cells prior to use for in vivo experiments (in b-c). b, Liver metastasis disease burden (in mm^3) following tail vein injection of Pip4k2c^KO/Insr^shIR without dox, GDC-0941 alone, dox-induced Insr knockdown alone, and combination of dox-induced Insr knockdown and GDC-0941. c, Lung metastasis burden (metastasis count) in corresponding animals from (b); n=10 mice per group. d, Blood glucose levels (in mg/dL) measured in mice (n=7 mice per group) following treatment with indicated diet or drugs (30 minutes post-administration) or vehicle control. e, C-peptide levels (in pM) following treatment with indicated combinations of GDC-0941 with SGLT2 inhibition or animals fed with ketogenic diet (n=11 mice per group). f, Liver metastasis disease burden (in mm^3) following tail vein injection of Pip4k2c^KO cells with drug treatments (SGLT2i, GDC-0941), ketogenic diet or combinations thereof in indicated groups. g, Lung metastasis burden (metastasis count) in corresponding animals from (f); n=10 mice per group, mean± s.e.m.; h, Tumor growth of subcutaneously injected HCmel12 parental or Pip4k2c KO melanoma cell lines following no treatment, treatment with GDC-0941 alone, or combination of GDC-0941 plus SGLT2 inhibition; n=5 mice per group, mean± s.e.m. i, Tumor diameter at day 24 in indicated treatment and genotype groups from (h). Box plots denote the minima, maxima and median in (h). Statistical significance was determined using One-Way ANOVA Tukey’s multiple comparisons test for (b-h). Data are representative of three (b-g) and two (h, i) independent experiments. Significance levels as indicated. Combination interventions reduce LM burden We reasoned that abolishing the PI3K-i-induced forward loop could reduce liver metastatic burden. To test this, we treated animals with an inhibitor of sodium-glucose co-transporter 2 (SGLT2i), that inhibits glucose re-uptake in the kidney, thereby, preventing spikes in insulin levels, or we fed animals a ketogenic diet that depletes glycogen storage (Methods). Peak blood glucose and plasma C-peptide levels were strongly reduced when combining GDC-0941 with SGLT2i or a ketogenic diet ([261]Fig. 5d,[262]e). In Pip4k2c^KO LM-bearing animals, combination of either SGLT2i or a ketogenic diet with GDC-0941 significantly reduced liver metastasis compared to animals receiving GDC-0941 alone ([263]Fig. 5f). Notably, combination of ketogenic diet with GDC-0941 also significantly reduced lung metastatic burden, while SGLT2i plus GDC-0941 selectively targeted liver-metastatic disease ([264]Fig. 5g), suggesting that addition of SGLT2i and a ketogenic diet to PI3K-i could circumvent GDC-0941-induced increase in LM. Importantly, compared to parental cells, loss of Pip4k2c had no differential impact on growth of subcutaneously implanted tumors or responses to GDC-0941 or GDC-0941 plus SGLT2i ([265]Fig. 5h,[266]I, [267]Extended Data Fig. 6b). This is consistent with the initial CRISPR-Cas9 screen and suggests PI3Ki-based therapeutic combinations are critical for abrogating insulin-mediated circuits that are most pronounced in LM ([268]Fig. 6). Figure 6. Schematic of Liver metastatic organotropism. Figure 6. [269]Open in a new tab Model illustrating increased liver metastatic burden in animals injected with Pip4k2c^KO cells, (1) effects of systemic PI3K inhibition, and strategies to overcome compensatory feedback loop and increased liver metastatic burden with either (2) ketogenic diet or (3) SGLT2-inhibition. Discussion Not all metastatic sites are created equal. Compared to other sites, metastasis to the liver confers a poorer prognosis across multiple cancer types and reduced responses to modern cancer therapies^[270]4,[271]31,[272]32. Mechanisms that determine a cancer cell’s metastatic fate towards one organ site over another are poorly understood and are not sufficiently accounted for by a model of serial, divergent acquisition of somatic mutations^[273]33,[274]34. The fact that cancer types with different oncogenic dependencies and distinct lineages exhibit a shared preponderance for metastasis to a specific organ - for example, liver metastasis in both melanoma and colorectal cancer, suggests that target-organ specific adaptations occur and may be determined by the required metabolic requirements and phenotypic plasticity^[275]35. Dissecting the molecular underpinnings of LM has been challenging due to limited pre-clinical models that recapitulate the liver-metastatic cascade in the presence of an intact immune system. Recent studies using intra-hepatic or portal/splenic vein injections of cancer cells revealed important but narrow aspects of LM biology and growth^[276]36,[277]37. Here, we leveraged a syngeneic melanoma model that has the potential to develop LM upon tail vein injection, thus, recapitulates several bottle-necks in the LM cascade. We performed functional screens and find that cancer cell intrinsic loss of Pip4k2c promotes melanoma cell metastasis by co-opting an insulin-rich liver milieu via activation of the PI3K/AKT pathway ([278]Fig. 6), without altering primary tumor growth or their metastatic potential to other organ sites (e.g., lung). Importantly, this mechanism may apply for both routes of dual blood supply to the liver, as we demonstrate increased LM via systemic circulation in our melanoma model and a prostate cancer model, and via portal vein, using a colorectal cancer model implanted into the colonic submucosa. Recent studies demonstrate that hyperinsulinemia, either in the context of insulin resistance or in response to systemic PI3K inhibition, increases tumor growth of subcutaneously implanted patient-derived xenograft models through inhibition of host PI3K^[279]28,[280]38,[281]39. Our study extends on this observation and shows that insulin-mediated tumor growth disproportionally occurs in LM over primary tumors or other metastatic sites, in line with higher insulin levels in the liver. These effects could be abrogated by reducing the insulin gradient between the liver and other organs through systemic delivery of insulin. Furthermore, combination of systemic PI3K inhibition with either inhibition of SGLT2 or a ketogenic diet could disrupt the effects of host-mediated increase in insulin levels and reduce LM, while having little or no effect on lung metastasis burden. These findings have potential clinical relevance in specific disease contexts. For example, patients with hormone receptor positive breast cancer, for which PI3K inhibitor alpelisib is FDA-approved and ongoing studies test the benefit of adding SGLT2 inhibition or a ketogenic diet ([282]NCT05090358), may particularly benefit from such strategies, when LM are present. While activating mutations of PIK3CA are frequent in breast cancer and serve as the basis for the use of PI3K inhibitors in breast cancer, our study suggests that this oncogenic dependency may not be necessary to select patients toward PI3K inhibitor-based combination treatment strategies, thus, expanding the potential pool of patients who may benefit from such therapies. Our study has potential limitations: Fist, loss or reduced expression of Pip4k2c likely represents one example of resulting hypersensitization to insulin, and may have effects beyond regulation of this pathway that we did not measure^[283]40,[284]41. Our initial screen only targeted kinases, thus, more extensive unbiased approaches are necessary to characterize the entire landscape of potential drivers of LM development. Second, analysis of human data showed that PIK3CA mutations are infrequent in melanoma, but enriched in melanoma LM, thus, suggesting that genomic activation of the pathway may directly drive LM in a portion of patients, independent from regulation by Pip4k2c or its isoforms. Importantly, even in the presence of PIK3CA mutations, insulin is still required to activate the pathway. Third, transcriptomics analyses of human data may suffer from contamination by normal hepatocytes in LM, and respective parenchymal and infiltrating cells in other analyzed organ sites. However, it is unlikely that this factor had a major effect, as we observed similar pathway enrichment in mouse metastases profiled by single-cell RNA-sequencing, where analyses were restricted to malignant cells. Lastly, further work is required to determine the role of niche cells and the tumor-microenvironment in liver-metastatic organotropism. Methods Ethics statement The research performed in this study complies with ethical regulations. Human specimen research was approved by the Institutional Review Board at Columbia University Irving Medical Center (#AAAO5706). All mouse experiments were performed under IACUC approved animal protocols at Columbia University (AABE6570) and Harvard Medical School (AABQ9616). The maximum subcutaneous tumor size permitted by both IACUCs is 20mm and was never exceeded. Source data is provided for all in vivo experiments. Mice 6–8 -week-old C57BL/6J (Charles River: 027), NOD/SCID/IL2gR (NSG) (JAX: 005557) and/or BALB/c (JAX: 000651) female mice were obtained from Charles River and/or Jackson Labs. Mice received a normal chow diet (PicoLab Rodent 20 5053 laboratory Diet St. Louis, MO) or a ketogenic diet (Thermo-Fisher AIN-76A) where indicated, with free access to drinking water. Diet composition is described in the [285]Supplementary Table 7. Mice were housed under pathogen-free conditions. Sex was not considered in the study design. Cell culture Mouse melanoma cell line HCmel12 was kindly provided by Prof. Thomas Tüting, Magdeburg, Germany. Melanoma cells were grown in ‘complete’ RPMI-1640 medium (Gibson Bioscience) as described previously^[286]13. RPMI 1640 supplemented with 10%FBS (Gibco^™ 10437028), 10mM non-essential amino acids, 1mM HEPES (all from Life Technologies), 20 μM 2-mercaptoethanol (Sigma). Mouse colorectal carcinoma cell line CT26 was cultured in RPMI 1640 medium supplemented with 10% FBS. A375 and HEK293 cells were purchased from ATCC and grown in DMEM supplemented with 10% FBS. Cell lines were routinely tested for mycoplasma contamination using Lonza MycoAlert Kit (Cat. # LT07–318). Generation of a Cas9-expressing HCmel12 melanoma cells Cas9 lentivirus was generated from HEK-293T cells transfected with pLX-311Cas9v.2 plasmids (Addgene #118018) and packaging plasmids pMD2.G (Addgene, #12259) and psPAX2 (Addgene, #12260) using TransIT-LT1 Transfection reagent (Mirus). HCmel12 melanoma cells were transduced with Cas9 lentiviral particles followed by blasticidin selection (1μg/ml) for 48 hours. To assess Cas9 cutting efficiency in HCmel12-Cas9 expressing cells, cells were transduced with pXPR_011 (Addgene #59702; gift from John Doench and David Root). Lentivirus expressing EGFP, puromycin resistance and EGFP-targeting sgRNA. Cas9 activity activity was estimated based on the fraction of EGFP-negative cells 7 days later using FACS Canto flow cytometry (BD Biosciences) and analyzed with FlowJo software (TreeStar, v10 for Mac). The fraction of EGFP-negative was used to estimate Cas9 efficiency. Propagation of Kinome sgRNA libraries and Lentivirus production Mouse Kinome CRISPR Knockout Library (Brie) was purchased from Addgene (Addgene, #75316), that includes 2,852 sgRNAs targeting 713 kinase genes and 100 control non-targeting sgRNAs ([287]Supplementary Table 3). For library production, HEK293T cells were seeded in DMEM + 10% FBS in T175 flasks one day prior. At 80% confluency, cells were transfected with the Brie library and the packaging/envelope plasmids pMD2.G (Addgene, #12259) and psPAX2 (Addgene, #12260) using TransIT-LT1 Transfection reagent (Mirus). After 6 hours media was replaced with DMEM + 20% FBS, and virus was collected after 48 hours, filtered, and stored at -80 °C. In vivo CRISPR-Cas9 screen HCmel12-Cas9 melanoma cells were transduced at a MOI < 0.1 with Brie lentivirus at ≥1000 cells/construct per infection replicate (~3x10^7 pre replicate). Cells were spin-infected at 1000 x g, 37°C for 2 hours and incubated for 16 hours followed by a media change. On day 2 post transduction the cells were put into Blasticidin selection (1 μg/ml) for 7 days and Puromycin (1 μg/ml) for the remainder of the experiment. On day 14, 3x10^6 of library-transduced HCmel12-Cas9 cells were injected into the right flank or lateral tail vein of the mice. In parallel cells were kept in culture for 21, 28, 35, 42, 49,52 days. Tumor growth and signs of sickness were monitored over time. Mice were euthanized after 4 weeks and tumors and metastases were evaluated for metastasis presence, lesions were dissected for gDNA isolation, sgRNA amplification, and sequencing. Genomic DNA was extracted using Qiagen DNeasy Blood & Tissue Kit (69506, Qiagen). sgRNA amplification and sequencing library construction were performed as described previously^[288]15, and sgRNA library was sequenced on a Illumina HiSeq2500 Instrument. CRISPR–Cas9 screens were analyzed and visualized using software packages MAGeCK and MAGeCKFlute R (v4.0.3)^[289]42. Generation of KO, knockdown and gene-overexpressing cell lines Individual tumor cell KOs were generated using Cas9 ribonucleoproteins (RNPs) nucleofection as described previously^[290]43. Target sequences were designed using CHOP-CHOP tool ([291]https://chopchop.cbu.uib.no) or sequences were derived from the original kinome library (sequence information in [292]Supplementary Table 8). For insulin receptor knockdown experiments doxycycline-inducible shRNA was used as described previously^[293]28 (Sequence information in Extended Data Table 8. Briefly, knockdown of insulin receptor was achieved using a doxycycline-inducible shRNA strategy. For generation of miR-E shRNAs, 97-mer oligonucleotides were purchased coding for predicted shRNAs using Splash RNA, [294]http://splashrna.mskcc.org/27. Oligonucleotides were PCR amplified using the primers miRE-Xho-fw (5′-TGAACTCGAGAAGGTATATTGCTGTTGACAGTGAGCG-3′) and miRE-Eco-rev (5′-TGAACTCGAGAAGGTATATTGCTGTTGACAGTGAGCG-3′). To generate full-length and N-terminal mutant Pipk4k2c overexpression constructs, open-reading frames were cloned into pLX305 using Gateway cloning ([295]Supplementary Table 8). Human pDONR223-PIP5K2C (Addgene, #23450) and N-terminal mutant human PIP4K2C, ORF were cloned into pLX307 using Gateway cloning and Gibson cloning, respectively. All vectors were sequence verified using Sanger sequencing followed by whole plasmid sequencing. Proliferation assay 1000 cells were seeded in 96-well plates. After 1, 2, or 3 days of proliferation, cells were fixed in 4% PFA and stained with DAPI. Timepoint zero was used as a reference control. Images were acquired on IVOS microscope, and analysis was performed using Cell Profiler software ([296]https://cellprofiler.org). Transwell migration assay Migration was assessed using transwell assays. 2×10^4 melanoma cells pre-treated with insulin were placed in the upper compartment of a transwell chamber with 8.0 μm pore size (Corning, 3422) containing 300 μl (top) and 700 μl media in the lower chamber. Cells were led to adhere and transmigrate through the membrane at 37 °C 5% CO2. Wells treated with no insulin served as controls. After 16 hours, transmigrated cells on the lower surface of the membrane were stained with CFSE, fixed with 4% paraformaldehyde and the mean number was counted in 5 high power fields using Inverted Microscope (Olympus). Immunoblots Cells were lysed in 1x Laemmli buffer (10,000 cells per μl) followed by 5min incubation at 95°C. Lysates were separated by 10% SDS–gel electrophoresis and transferred to a PVDF membrane (IB24001, Fisher Scientific). Membranes were blocked for 1 h (11921673001, Roche blocking buffer) in Tris-Buffered Saline, pH 7.6 (TBS, S196830–2, Agilent Technologies) and probed with primary antibodies overnight at 4°C followed by 1h incubation in secondary antibodies at RT. Proteins were detected using the Odyssey CLx Imaging system (LI-COR Biosciences). Antibody information is provided in the Reporting Summary of the paper. Membranes were stripped using Stripping Buffer (46430, Thermo Scientific). Full blots corresponding to the immunoblots shown in the main and [297]supplementary figures are provided as a Source File. RNA isolation, RNA sequencing and analysis 2x10^6 parental and Pip4k2c^KO HCmel12 melanoma cells were plated in 6-well plates FBS-free media overnight, with three biological replicates per condition, total 12 samples. The next day cells were treated with insulin (125ng/ml) for four hours ([298]Supplementary Table 4). RNA was extracted using Qiagen kit (74136) and sequenced on a HiSeq2500 (Illumina). Transcripts were quantified with Salmon^[299]44 using the mouse reference genome release GRCm38.97. The tximeta R package (v.4.3)^[300]45 was used to import transcript-level counts and aggregate them by gene. DESeq2 package was applied to generate normalized gene counts. Gene set enrichment analysis (GSEA) for pathway genes was identified using BROAD java GSEA software with normalized gene counts as input ([301]https://www.gsea-msigdb.org/gsea/index.jsp). In vitro drug treatment Cells were washed 1x in PBS, placed in FBS-free media overnight, and treated for 1h with PI3K inhibitors (Pictilisib/GDC-0941 (S1065) and Alpelisib (S2814) from Selleck Chemicals) as indicated with insulin for 15 min before collection for immunoblotting. To induce insulin receptor knockdown cells were treated with 1μg/ml doxycycline (D9891–5G, Sigma) for 48 hours, washed 1x with PBS, and subjected to signaling studies. Animal metastasis and drug treatment studies in vivo 5x10^5 HCmel12 mouse melanoma cells, 1x10^5 A375 human melanoma cells were injected into the lateral tail vein. Mice were monitored over three to four weeks. When mice showed signs of sickness, 20% of weight loss, impaired activity, hunching, and limb paralysis, animals were euthanized, and organs were harvested for evaluation on metastasis presence. For in vivo drug treatment studies, Pictilisib (GDC-0941, HY-50094) and Canagliflozin (JNJ 28431754, HY-10451) were purchased from Medchem Express (Monmouth Junction, NJ). The day after tumor cells injection mice were given vehicle control or treated (daily, 5 out of 7 days) with GDC-0941 (100 mg/kg) or SGLT2i (6 mg/kg) prepared in ‘CMC’ solution, 0.5% carboxy-methyl-cellulose sodium (CMC, C9481–500G, Sigma) in distilled water supplemented with 0.2% Tween-80 (BCBW1801, Sigma) for the duration of the experiment unless otherwise stated. To maintain Insr knockdown in vivo, mice bearing Pip4k2c^KO cells with dox-inducible Insr shRNA were administered 3 mg/kg of doxycycline (Sigma, D3072) intraperitoneally daily, for three to four weeks. Ketogenic diet was started 10 days prior to tumor cell injection where indicated. In experiments combining GDC-0941 and SGLT2 inhibitor (Canagliflozin), the latter was administered 60 min prior to GDC-0941^[302]28 administration. 2x10^5 tumor cells were injected subcutaneously and tumor growth was monitored over time. When tumors were palpable mice were treated with GDC-0941 (100mg/kg) alone or in combination with SGLT2i (6mg/kg), 3 times a week until the end of the experiment. Mice were euthanized and tumor growth was evaluated. Endoscopy-guided orthotopic tumor transplantation and injections Orthotopic injections of CT26 cells was performed as described previously^[303]46. Briefly, cell lines were split 2 days prior. On the day of injection, cells at 80–90% confluency were harvested by TrypLE Express enzyme (Gibco, 12604013). Tumor cells were resuspended in PBS + 10% Matrigel at 2x10^6/ml. 100μl was injected into the colon sub-mucosa of each BALB/c recipient mouse by optical colonoscopy using a Hamilton syringe (7656–1) and a custom 33G needle (Hamilton, custom made similar to 7803–05, 16”, Pt 4, Deg 12). Successful injections were confirmed by observing large bubbles in the colon mucosa. After 14 days mice were euthanatized and organs evaluated for metastasis presence. Optical colonoscopy was performed two weeks later to assess tumor formation using a Karl Storz Image 1 HD Camera System, Image 1 HUB CCU, 175-Watt Xenon Light Source, and Richard Wolf 1.9mm/9.5 Fr Integrated Telescope (part number 8626.431). One injection was performed per mouse. Implantation of Insulin Pads in NSG mice NSG mice were implanted with 2x3 mm slow-release insulin pellets (LinBit - LinShin, Canada) on the dorsum under the skin. Under isoflurane anesthesia, mice were shaved, the surface of the skin treated with a 70% isopropyl alcohol and a lidocaine-based cream (e.g. Emla). The insulin pellet was inserted using 12G trocar and the skin opening is sealed with a drop of Vetbond. After a day, 1x10^5 tumor cells were injected via lateral tail vein and tumor metastatic burden was evaluated after four weeks. Blood glucose, C-peptide and tissue insulin measurements 10 μl of blood was taken from the tail of mice before treatment and at indicated time points (15, 30, 60, 90, 120,180, 240 min) and blood glucose was measured using a Prodigy AutoCode Glucometer. At the end point >100 μl of blood was drawn from the mice into heparin coated tubes (07980, Stem Cell Technologies). Blood was centrifuged (10,000g for 10 min at 4 °C), and plasma was stored at −80 °C. Mouse and human C-peptide levels in plasma and mouse insulin levels in liver and lung tissue were measured using commercial ELISA kits (Mouse C-peptide ELISA: 80-CPTMS-E01 (ALPCO); STELLUX Chemiluminescence Human C-peptide ELISA: 80-CPTHU-CH01 (ALPCO); Salem, NH; Insulin: Invitrogen cat. #EMINS). Tissue collection Liver and lung tissue were collected immediately after mice were sacrificed, and snap-frozen in liquid nitrogen. For tissue protein isolation, 5mg of powdered liver and lung tissue were homogenized in 300μl Mammalian Protein Extraction Reagent (catalog number: 78501) buffer with proteinase inhibitors (catalog number: 1861281) and incubated at 4 °C for 2 hours with rotating, followed by 20min spin at 20.000g, 4 °C. Supernatant was collected and stored at -80°C. FDG-PET C57/BL6 mice were injected with Pip4k2c^KO melanoma cells. After 3 weeks mice were scanned for liver metastasis presence using PET with [F18] fluorodeoxyglucose (FDG) tracer. At the time of peak blood insulin feedback 60 min post-GDC-0941 injection animals were injected IV with FDG. 60 min later mice were placed into a 4-mouse bed and 30 min static PET images were acquired using a PET scanner (Siemens, Munich, Germany) followed by micro CT (MILabs, Houten, Netherlands) on the same bed for anatomical references. Regions of interests (ROI) were