Main

Recent work has leveraged the connection between genetic and epigenetic interactions regulating the development of tumorigenic phenotypes to identify cancer-specific vulnerabilities. Recurrent loss-of-function deletions are prevalent genomic alterations, cause frequent inactivation of tumour suppressor genes and are important driver events in tumorigenesis1. Although these deletions offer functional and fitness advantages to cancer cells, they make them vulnerable due to the collateral deletion of essential genes in chromosomal proximity2,3,4,5,6,7. Essential passenger gene deletions in cancer cells can concomitantly lead to dependence on paralogues that maintain a similar function. While the loss of genes that encode metabolic enzymes may block a metabolic pathway, redundancy in the metabolic circuitry and plasticity of cancer cells allows them to utilize alternative pathways not predictable from first principles. The emergence of these context-specific compensatory pathways gives rise to the concept of collateral lethality. Although frameworks have been developed recently to uncover gene associations for co-essentiality mapping and gene essentiality8,9,10,11,12, no current methodology can systematically uncover collateral lethal genes for deleted passenger metabolic genes. Identification of paralogues is crucial to exploiting the vulnerabilities of cancer cells while sparing normal cells.

Previous approaches identified these paralogous metabolic genes by considering the functional compensation occurring through enzyme isoforms2,3,4,13. While current approaches—including genetic loss-of-function screens—can reveal paralogues based on gene essentiality, there is a pressing need for a methodology that can (1) dissect the differential metabolism within subcellular compartments, (2) incorporate redox and biosynthetic balances maintained through interorganelle shuttling and (3) use metabolic fluxes to map genetic information with phenotypic biological functions. This comprehensive methodology will not only lead to robust identification of paralogues but also highlight the interconnectedness of metabolic networks by considering multiple cellular objectives beyond just proliferation.

We have developed an approach, defined as collateral lethal genes identification via metabolic fluxes (CLIM), that identifies personalized metabolic targets in cancers with distinct mutational signatures using a combination of data mining, machine learning and multi-objective metabolic flux analysis techniques. To determine putative frequent heterozygous and homozygous deletions of metabolic enzyme-encoding genes at the global genomic landscape level, we analysed copy number alteration (CNA) data from The Cancer Genome Atlas (TCGA) pan-cancer dataset of >10,000 patients with GISTIC2.0 (ref. 14) and uncovered the presence of 80 frequently deleted chromosomal regions (Fig. 1a). Despite, the genetic heterogeneity of tumours from 32 sites of origin, these data strongly indicate that certain genomic loss events are common among cancer alterations and are conserved across primary sites14. Interestingly these focal deletions contain at least 49 metabolic genes, which further supports our hypothesis of prevalent loss of metabolic genes in cancers (Fig. 1a). By performing a similar analysis on CNA from tumours categorized by the site of origin, we revealed a similar pattern of frequently occurring chromosomal deletions containing metabolic genes (Extended Data Fig. 1a,b). Solid tumours showed higher numbers of chromosomal deletions compared with acute myeloid leukaemia (AML) samples, and the number of potentially lost metabolic genes ranged from 26 in AML to 212 in invasive breast carcinoma (Extended Data Fig. 1b). Interestingly, cross-referencing these metabolic genes to a database of genetic paralogues uncovered that almost half of the deleted metabolic genes did not have genetic paralogues (Extended Data Fig. 1c). This further highlighted the need to identify potential collateral lethal pathways in tumour metabolic networks. Further, from the high-grade serous ovarian carcinoma (HGSOC) TCGA dataset with prevalent chromosomal alterations we observed 60 frequent deletions containing >170 metabolic genes (Fig. 1b). We devised an integrated workflow that utilizes genomic and transcriptomic profiles of cancer patients to identify metabolic deletions, reconstructs genome-scale metabolic models (GSMMs), performs cellular objectives-based metabolic flux analysis and uncovers compensatory metabolic pathways for collateral lethal targeting (Fig. 1c and Extended Data Fig. 1d). Interestingly, the most prevalent deletion on the 19p13.3 locus in HGSOC was identified not only by the GISTIC2.0 algorithm but also by our machine learning approach, which stratifies patients based on oncogenic mutations and CNA (Fig. 1c,d). The 19p13.3-deleted region contains the tumour suppressor LKB1, which is mutated or deleted in Peutz–Jeghers syndrome and several cancers and is involved in the tumorigenesis of cervical and ovarian cancers15,16, with homozygous loss in 5% and heterozygous loss in up to 30% of HGSOC tumours from TCGA17.

Fig. 1: CLIM: a systems, multiomics and machine learning integrated-precision oncology platform that identifies collateral lethal metabolic targets.
figure 1

a,b, Circos plots showing average genomic copy number gain and loss, most notable focal deletions in chromosomes 1–22 (identified by GISTIC2.0) and number of total genes and metabolic gene symbols within deleted regions for (a) the TCGA Pan-Cancer patient dataset (n = 10,104) and (b) the TCGA HGSOC dataset (n = 606). c, Schematic describing the workflow used to (1) identify focal deletions in patients with particular genetic profiles, (2) determine the occurrence of metabolic vulnerabilities due to collateral loss of metabolic genes, (3) perform multi-objective, genome-scale metabolic flux analysis to reveal compensatory metabolic pathways that are collateral lethal targets and (4) test the validity of collateral lethal metabolic genes. d. Heatmap of scores obtained via machine learning NNMF algorithm to stratify ovarian tumours based on two molecular features: mutation status and CNA (specifically large-scale deletions). Each patient cluster (columns, vertical bar chart) is defined by a combination of molecular features wherein NNMF-derived cluster scores quantify the importance of molecular features (rows) in defining a patient cluster. Molecular features are sorted in descending order of the average of their scores (horizontal bar chart) across all patient clusters, representing the most important to the least important molecular features in ovarian tumours. CNA along chromosome 19 highlights copy number loss in 19p13.3 locus for five patient strata (clusters 3, 11, 12, 23 and 32), all of which have a high cluster score for the 19p13.3 deletion. CNA along chromosome 19 for cluster 21, which is defined by other molecular features and has the lowest cluster score for 19p13.3 deletion, shows no copy loss in the locus (CNA is approximately 0).

Source data

To delineate patient clusters with distinguishable chromosomal deletions and mutations at a population level on the HGSOC dataset, we performed unsupervised clustering analysis based on molecular features using non-negative matrix factorization (NNMF). Putative oncogenic mutations had the highest scores in defining NNMF clusters; remarkably, 19p13.3 CNA was among the top cluster-defining molecular features (Fig. 1d). 19p13.3 deletion was a distinguishing feature for five NNMF clusters, and contrasting the average CNA for these patient clusters (clusters 3, 11, 12, 23 and 32) showed consistent copy loss in 19p13.3 whereas other loci of chromosome 19 had no discernible commonalities. The depth of 19p13.3 deletion in these clusters is further highlighted when comparing the CNA of these five clusters with that of cluster 21, which had no CNA of the 19p13.3 locus. Ubiquinol-cytochrome c reductase, complex III subunit XI (UQCR11), a gene that encodes a subunit of complex III of the electron-transport chain (ETC), lies within this locus. Furthermore, copy loss of UQCR11 strongly correlates with reduced messenger RNA expression of UQCR11 (Extended Data Fig. 1e). Because UQCR11 has no known genetic paralogue, its loss would indicate mitochondrial energy metabolism dysfunction. Although the systemic pathological role of complex III dysfunction in healthy cells is rare and undercharacterized18, it has been shown to impair oxidative phosphorylation (OXPHOS) in breast cancer19,20 and contributes to destabilization of complex I21. Because OXPHOS is an essential source of ATP for rapidly proliferating cancer cells, mitochondrial dysfunction drives metabolic rewiring to meet high bioenergetic demands22,23. This led us to hypothesize that ovarian cancer cells with the 19p13.3 deletion and ETC impairment would shift their reliance onto other metabolic pathways to maintain their energy requirements and NAD+/NADH balance, revealing a collateral lethal metabolic target specific to UQCR11-null ovarian cancer.

To capture the plasticity of cancer metabolism, we relied on comprehensive GSMMs to dissect the effect of UQCR11 loss and ETC impairment in HGSOC. Multiple cellular objectives that compete for metabolic resources reflect the adaptability of cancer cells to varied intrinsic and extrinsic pressures. Herein, we selected competing objectives of growth (biomass production) and survival (ATP production) (Supplementary Information 1). We employed our multi-objective metabolic flux analysis (MOMFA) algorithm on reconstructed GSMM for 19p13.3-deleted ovarian cancers to reveal the compensatory metabolic pathways employed by cancer cells after loss of UQC11-mediated reactions (Fig. 2a,b and Extended Data Figs. 2 and 3a–d). Those compensatory pathways that were ubiquitously active across the spectrum of adaptability were considered as viable candidates. From these, pathways that detrimentally affected 19p13.3-deleted cells when inhibited in silico while also having a minimal effect on cells without 19p13.3 deletions were ranked higher by CLIM (Supplementary Information 2). The top chosen candidate pathways—catalysed by enzymes encoded by methylenetetrahydrofolate dehydrogenase, MTHFD1/2 and phosphoserine aminotransferase PSAT—revealed an intriguing rewiring in cells with UQCR11 deletion (Extended Data Fig. 3). Surprisingly, despite substantial rewiring of mitochondrial one-carbon metabolism, the predicted nucleotide synthesis fluxes were minimally affected (Fig. 2b). MTHFD2 was predicted as the most critical component of one-carbon metabolism and emerged as a paralogous pathway that is contextually active in cells with UQCR11 deletion (Extended Data Fig. 3a,d and Supplementary Information 1 and 2). Whole-genome sequencing data from the Cancer Cell Line Encyclopedia (CCLE) helped identify four ovarian cancer lines that would represent copy loss and diploid UQCR11 (Extended Data Fig. 4a). CAOV3, HEYA8 and OVCAR8 with heterozygous UQCR11 deletion (hereafter termed UQRC11-null) and SKOV3 with diploid UQCR11 (hereafter termed UQCR11-intact) were employed for evaluation of collateral lethal targeting in vitro. SKOV3 expressed UQCR11 at the transcript and protein levels while CAOV3, HeyA8 and OVCAR8 had undetectable UQCR11 protein expression (Fig. 2c,d), validating our in vitro models.

Fig. 2: Multi-objective metabolic flux analysis identifies MTHFD2 as a collateral lethal target in UQCR11-null ovarian cancers with in vitro, clinical and pan-cancer implications.
figure 2

a, Workflow elucidating multi-objective flux analysis algorithm comparing flux distributions of cancers with and without deletions for identification of collateral lethal pathways. b, Multi-objective metabolic Pareto frontier and metabolic fluxes of MTHFD1, MTHFD2, RNA and DNA synthesis pathways in UQCR11-null and -intact models. Radar plots represent Pareto-optimal flux distributions from maximal biomass to maximal ATP maintenance (counterclockwise). c, mRNA expression of UQCR11 and MTHFD2 in parental ovarian cancer cell lines. Data presented as mean ± s.d. (n = 6), analysed using two-tailed Student’s t-test. d, Protein expression of MTHFD2 and UQCR11. Representative immunoblots with β-actin as loading control. e, Cell viability after dox-induced shMTHFD2 knockdown. Data representative of two independent experiments and presented as mean ± s.d. (n = 3), analysed using two-way analysis of variance (ANOVA) with Dunn’s correction. f, Viability of parental ovarian cancer lines treated with MTHFD2 inhibitor, DS18561882. Data representative of two independent experiments and presented as mean ± s.d. (n = 4). Data were fit to a three-parameter, variable-slope curve. g, Viability of isogenic SKOV3 lines expressing shScramble or shUQCR11, treated with MTHFD2 inhibitor. Data combined from two independent experiments and presented as mean ± s.d. (n = 6). Data were fit to a four-parameter, variable-slope curve. h, One-carbon and mitochondrial metabolism genes that predict progression-free survival in TCGA patients with ovarian cancer, based on 19p13.3 deletion status using RSF. i, Correlation of mRNA expression of MTHFD2 and UQCR11 in 20 cancer types from TCGA Pan-Cancer dataset. Error bars represent 95% confidence intervals of correlation coefficients estimated with Spearman’s rank correlation test. UQCR11 and MTHFD2 mRNA expression in TCGA UCEC, TCGA UCS tumours and CCLE endometrial cancer lines (n = 612, top inset). MTHFD2 and UQCR11 mRNA expression and dependency scores of MTHFD2 (n = 18, bottom inset). kidney PC, kidney papillary carcinoma; kidney CC, kidney clear cell carcinoma; stomach ade, stomach adenocarcinoma. j, Overall survival of patients with UCEC and UQCR11 copy loss (below median) with high or low MTHFD2 mRNA expression (top and bottom quartiles), compared with patients with no copy loss (above median). Data analysed pairwise by log-rank test. k, Overall survival of patients with UCEC and low UQCR11 (bottom quartile) expression and high (top quartile) expression of MTHFD2 compared with patients with low UQCR11 and MTHFD2 (bottom quartile) expression. Data analysed by log-rank test. HR, hazard ratio.

Source data

MTHFD2 short hairpin RNA transfection decreased cell viability in a dose-dependent manner in UQCR11-null OVCAR8, CAOV3 and HeyA8 cell lines but had no effect on the UQCR11-intact SKOV3 line (Fig. 2e and Extended Data Fig. 4b). These observations were corroborated with doxycycline (dox)-inducible shRNA-mediated stable knockdown of MTHFD2 in UQCR11-intact SKOV3 and UQCR11-null OVCAR8 lines (Extended Data Fig. 4c,d). Specifically, dox induction of shMTHFD2 decreased the cell viability of OVCAR8 but had no effect on SKOV3 cells (Extended Data Fig. 4e). This collateral lethal targeting effect was also confirmed using DS18561882, a highly potent and selective MTHFD2 inhibitor24 (Fig. 2f), with UQCR11-null cells showing six- to 20-fold lower concentration causing 50% cell growth inhibition (GI50) compared with UQCR11-intact cells. Furthermore, in an isogenic model developed by stably expressing shUQCR11 in UQCR11-intact SKOV3 cells, GI50 concentrations were notably reduced in SKOV3 cells with UQCR11 knocked down, indicating a strong functional collateral lethal link between UQCR11 deletion and targeting MTHFD2 (Fig. 2g and Extended Data Fig. 4f). To discover the metabolic link between UQCR11 and MTHFD2, we first assessed the metabolic influence of UQCR11 deletion on ovarian cancer cells. Notably, UQCR11 knockdown did not affect cell viability but significantly reduced total cellular oxygen consumption rate (OCR) (Extended Data Fig. 4g,h and Supplementary Fig. 1). Further, basal OCR in UQCR11-null parental ovarian cancer lines was reduced compared with UQCR11-intact cells (Extended Data Fig. 4i). At the metabolic level, NAD+ levels were significantly lower and NADH levels trended lower in UQCR11-null cells compared with UQCR11-intact, in both parental and isogenic systems (Extended Data Fig. 4j,k). Furthermore, ETC impairment in UQCR11-null tumours was possibly exacerbated by the loss of NDUFS7, which lies in the same locus as UQCR11 (Extended Data Fig. 5a,b). To recapitulate a functional impairment of complex I, we inhibited its activity in UQCR11-intact SKOV3 cells with the complex I inhibitor, rotenone, and treated them with MTHFD2 inhibitor. Notably, pharmacological co-inhibition of complex I and MTHFD2 had a synergistic effect on the viability of SKOV3 cells, indicating the existence of a collateral lethal link between ETC impairment and MTHFD2 activity (Extended Data Fig. 5c). Additionally, re-establishing complex III activity in UQCR11-null HeyA8 cells by ectopically expressing alternative oxidase, AOX, from Ciona intestinalis25,26,27 and overexpressing UQCR11 partially rescued these cells from MTHFD2 inhibitor-mediated cell death (Extended Data Fig. 5d–g and Supplementary Figs. 2 and 3). These observations suggested that the lethality of targeting MTHFD2 in UQCR11-null cells was linked to ETC impairment, potentially via the imbalance of NAD+.

To establish the clinical relevance of MTHFD2 collateral lethality, random survival forest (RSF) models were trained for prediction of prognosis based on age and gene expression of metabolic pathways related to one-carbon metabolism, ETC and the Krebs (TCA) cycle (Supplementary Table 1). As expected, age best predicted mortality in all cohorts. Remarkably, MTHFD2 and MTHFR of one-carbon metabolism predicted poor prognosis in the 19p13.3-deleted cohort but not in the 19p13.3-intact cohort (Fig. 2h and Supplementary Fig. 4). These unsupervised machine learning methods predicting survival, therefore, provided further evidence on the importance of MTHFD2 and related metabolic genes in UQCR11-null ovarian tumours. We also sought to determine whether MTHFD2-centric rewiring occurs in other cancers with UQCR11 deletion. Remarkably, the strong negative correlation between UQCR11 and MTHFD2 transcripts indicated potential collateral lethal pairing in many cancers across the TCGA Pan-Cancer cohort (Fig. 2i); only gliomas showed a positive correlation between MTHFD2 and UQCR11. Interestingly, gliomas are outliers compared with other cancers beause low MTHFD2 expression is correlated with poor prognosis, alluding to MTHFD2 dependence unrelated to the collateral lethal pairing with UQCR11 (refs. 28,29). Notably, uterine cancers have a strong negative correlation, as seen in combined TCGA datasets for uterine corpus endometrial carcinoma (UCEC) and uterine carcinosarcoma (UCS), and uterine cancer lines from the CCLE (Fig. 2i, top inset). This correlation was strongest within UCEC samples (Extended Data Fig. 6a). In addition, deletion of the 19p13.3 locus was independently identified as significant in UCEC tumours using GISTIC (q-value = 2.3 × 10−21; Extended Data Fig. 6b). This deletion is notable given that previous observations in mouse models revealed that loss of LKB1 is linked to UCEC progression30,31. Furthermore, the consensus score of MTHFD2 dependency in endometrial cancer lines reported in the Cancer DepMap portal32 displayed a correlation that trended negatively with the ratio of MTHFD2 to UQCR11 expression, indicating MTHFD2 dependency in UCEC with low UQCR11 (Fig. 2i, bottom inset). Furthermore, we see that patients with uterine cancer and UQCR11 copy loss have a markedly poorer prognosis compared with those with diploid or amplified UQCR11 (Fig. 2j, yellow versus red and blue). Their poor outcomes are further exacerbated when tumours have both UQCR11 copy loss and high MTHFD2 gene expression compared with those patients with low MTHFD2 expression (Fig. 2j, red versus blue). Similarly, patients with tumours showing low UQCR11 gene expression and high MTHFD2 expression have poorer outcomes compared with patients with low expression of both UQCR11 and MTHFD2 (Fig. 2k), suggesting that tumours with 19p13.3 deletion but higher expression of MTHFD2 are more aggressive compared with those with lower MTHFD2 expression. Notably, a multilayer machine learning model within CLIM could predict UQCR11 copy loss and response to MTHFD2 inhibition in UCEC cancers with minimal mutation and gene expression data (Extended Data Fig. 6c–e, Supplementary Information 3 and Supplementary Tables 2 and 3). Interestingly, UCEC tumours with a TP53 mutation but not a PTEN mutation are predicted to display 19p13.3 loss whereas tumours with a TP53 and PTEN mutation, or no TP53 mutation, do not (Extended Data Fig. 6c). These data highlight that collateral lethal targets discovered by CLIM can capture targets across cancer types based on mutational and transcriptional information.

Although collateral lethal gene identification and its therapeutic targeting to control tumour growth have previously been demonstrated2,4,33,34, the regulation of systemic metabolic activity by paralogous metabolic pathways to achieve metabolic flux compensation has been overlooked. CLIM showed the unique behaviour of the mitochondrial MTHFD2 pathway by elucidating its reversed direction, producing NAD(P)+ with respect to the widely observed NAD(P)H-producing flux of mitochondrial SHMT2 and MTHFD2 reactions35,36,37. The MOMFA algorithm within the CLIM workflow indicated that this was due to the unique pleiotropic properties of MTHFD2 and its ability to generate NAD+ (Fig. 2b and Supplementary Information 1 and 2). We established the existence of metabolic compensation using isotope tracing with 2H deuterium-labelled substrates and validated MOMFA-predicted fluxes. Although the ETC substantively contributes to maintaining the NAD+/NADH ratio in mitochondria, three other pathways can also oxidize NADH to NAD+: aminomethyltransferases (AMT), glutamate γ-semialdehyde dehydrogenase (ALDH4A1) and MTHFD2 (Fig. 3a,b). In UQCR11-intact cells MOMFA predicted that, apart from ETC, AMT was the only pathway that would oxidize NADH to NAD+ across the spectrum of metabolic adaptations possible, represented by competing cellular objectives of growth and survival (Fig. 3a–c (teal areas) and Supplementary Information 1). MTHFD2 and ALDH4A1 fluxes contributed minimally to maintenance of mitochondrial NAD+ pools (Fig. 3b,c, blue and pink areas); in fact, net MTHFD2 flux was responsible for NAD(P)+ reduction to NAD(P)H (Fig. 3b,c, pink area). In contrast, ALDH4A1 fluxes were higher and AMT fluxes lower across the metabolic states between growth and survival in UQCR11-null cells compared with UQCR11-intact (Fig. 3b,d,e, teal and blue areas). The most remarkable change, however, was MTHFD2 flux, which compensated for UQCR11 deletion-induced NAD+ depletion by maintaining a net flux oxidizing NAD(P)H to NAD(P)+ across all possible optimal metabolic states between the two extreme cellular objectives (Fig. 3b,d,e, pink areas). This non-canonical flux of MTHFD2 (hereby referred to as oxidative MTHFD2 flux) was not observed in UQCR11-intact cells in silico, and further strengthened the candidacy of MTHFD2 as a collateral lethal target in UQCR11-null cells.

Fig. 3: Metabolic flux analysis with isotope tracing using deuterium-labelled substrates demonstrates metabolic flux compensation through a paralogue pathway.
figure 3

a, In UQCR11-intact cells, mitochondrial MTHFD2 maintains a flux in the reductive direction converting NAD(P)+ to NAD(P)H while ETC, ALDH4A1 and AMT maintain NAD+/NADH pools oxidatively. b, Pathways in mitochondria, apart from ETC, that rely on NAD+/NADH include AMT, ALDH4A1 and MTHFD2. Directions of flux correspond to schematics (a,d), and the associated sign of flux values in Pareto-optimal flux space (c,e) are displayed. c, Pareto-optimal fluxes of reactions oxidizing NADH to NAD+, MTHFD2, AMT and ALDH4A1, as predicted by MOMFA in UQCR11-intact cells, are plotted across the Pareto-optimal metabolic objectives of maximal growth and maximal survival. Fluxes are normalized to net pyruvate dehydrogenase (PDH) flux set to 100. d, In UQCR11-null cells, NAD+ depletion due to impaired ETC leads to MTHFD2 compensating for NAD+ depletion by maintaining a net flux in the oxidative direction, converting NAD(P)H to NAD(P)+. e, Pareto-optimal fluxes of reactions oxidizing NADH to NAD+, MTHFD2, AMT and ALDH4A1 as predicted by MOMFA in UQCR11-null cells are plotted across the Pareto-optimal metabolic objectives of maximal growth and maximal survival. Fluxes are normalized to net PDH flux set to 100. f, Stable-isotope tracing map elucidating the transfer of [2H] and enrichment of downstream metabolites from stable-isotope substrates 3-[2H]-glucose and 4-[2H]-glucose. g, Ratio of [2H]-serine/[2H]-NADH in UQCR11-null cells (OVCAR8, CAOV3 and HEYA8) relative to the same ratio in UQCR11-intact cells (SKOV3) after 24 h in 4-[2H]-glucose-labelled medium. Data combined from two independent experiments (n = 6), with mean ± s.d. estimated using error propagation and analysed by ordinary one-way ANOVA with Dunnett’s correction for multiple comparisons. h, Isotopic enrichment of M+1 [2H]-serine in UQCR11-null cells relative to M+1 [2H]-serine enrichment in UQCR11-intact cells after 24 h in 3-[2H]-glucose-labelled medium. Data representative of two independent experiments, presented as mean ± s.e.m. (n = 3) and analysed by ordinary one-way ANOVA with Dunnett’s correction for multiple comparisons.

Source data

Regulation of one-carbon metabolism, including MTHFD2, has been studied extensively in the context of regulation of cofactors and NAD(P)H;38,39,40,41 however, contextual directionality in the MTHFD2 pathway has not been reported. Parallel tracer experiments with three [2H]-labelled substrates (3-[2H]-glucose, 4-[2H]-glucose and 2,3,3-[2H]-serine) allowed uncovering of mechanistic underpinnings in this pathway in vitro (Extended Data Fig. 7a and Supplementary Information 4). Labelling of cytosolic NADH from 4-[2H]-glucose in all four cell lines is evident from the labelled lactate (Extended Data Fig. 7a, lactate M+1 in orange). Further, reaction catalysed by cytosolic malate dehydrogenase (MDH1) transfers [2H] from NADH to cytosolic malate, which is transported to the mitochondria where malate transfers [2H] back to NADH (Fig. 3f and Extended Data Fig. 7a, orange tracer pathway)36. Enrichment of total cellular malate indicates that there is indeed a transfer of [2H] between malate and NADH36,42 (Extended Data Fig. 7a, malate M+1 in orange). Only oxidative MTHFD2 flux can transfer [2H] from mitochondrial NADH to 5,10-methylene-THF, which is subsequently transferred from 5,10-methylene-THF to mitochondrial serine by SHMT2 (Fig. 3f, Extended Data Fig. 7a (orange tracer pathway) and Supplementary Information 4). Remarkably, the isotopic enrichment ratio of [2H]-serine to [2H]-NADH, which represents this oxidative MTHFD2 flux, was found to be threefold higher in UQCR11-null cells compared with UQCR11-intact cells (Fig. 3f,g). The distinct labelling of [2H]-serine from [2H]-NADH via oxidative MTHFD flux is possible only through mitochondrial MTHFD2 activity, which oxidizes NAD(P)H, while cytosolic MTHFD1 can use only NADPH. These data provide the first strong piece of evidence that MTHFD2 flux is reversed during ETC impairment, as in UQCR11-null cells. Furthermore, tracing with 2,3,3-[2H]-serine corroborated that flux through reversible MTHFD2 and SHMT2 is dominantly oxidative. Enrichment of M+1 serine increased over time in UQCR11-null cells (Extended Data Fig. 7b, light green tracer pathway) and concomitantly decreased in UQCR11-intact cells (Extended Data Fig. 7b, dark green tracer pathway). This, accompanied by decrease in M+3 serine and M+1 glycine (Extended Data Fig. 7b, dark green tracer), was possible only when MTHFD2 reduced 5,10-methenyl-THF to 5,10-methylene-THF while producing NAD(P)+, followed by demethylation of 5,10-methylene-THF to serine from glycine by SHMT2 in the direction of oxidative MTHFD2. From 3-[2H]-glucose tracer experiments, we followed the labelling of [2H]-NADPH in the cytosol to serine via MTHFD1, a prerequisite for nucleotide synthesis. Because enrichment of serine was similar across UQCR11-null and -intact cells, we concluded that nucleotide synthesis maintained by MTHFD1 was unaffected by UQCR11 deletion (Fig. 3f,h, Extended Data Fig. 7a (blue tracer pathway) and Supplementary Information 4). This was also predicted by MOMFA, where the net fluxes of DNA and RNA synthesis were similar between UQCR11-null and -intact conditions (Fig. 2b).

Importantly, UQCR11 copy loss potentially leads to hypomorphs where mitochondrial respiration is partially impaired, as observed from parental and UQCR11 knockdowns that survive despite lower OCR and reducing factors (Extended Data Fig. 4g–k). To dissect the emergence of oxidation of NADH through MTHFD2 during ETC inhibition, we treated UQCR11-intact SKOV3 cells with increasing doses of rotenone. Specifically, oxidative MTHFD2 flux first increased with increasing, yet low, rotenone doses but returned to original levels as doses were further increased (Fig. 4a). This flux is suppressed when cells are treated with the MTHFD2 inhibitor (Fig. 4a). Notably, in the low range of rotenone doses where oxidative MTHFD2 flux increased, there was a synergistic decrease in OCR in SKOV3 cells treated with rotenone and MTHFD2 inhibitor (Fig. 4a). At doses >62 nM, MTHFD2 inhibition did not detrimentally affect OCR despite a monotonic decrease in basal OCR with increasing rotenone concentrations (Fig. 4a,b). In the case of reductive MTHFD2 flux there was a modest increase at low rotenone doses, which became most pronounced at the highest doses (Fig. 4b). Expectedly, treatment with the MTHFD2 inhibitor greatly reduced reductive MTHFD2 flux (Fig. 4b). Cumulatively, these data support the emergence of oxidative MTHFD2 flux as a compensatory pathway for NAD+ production in cells with mild ETC impairment. This phenomenon subsequently yielded to reductive MTHFD2 flux under strong ETC inhibition, as previously demonstrated40. This non-monotonic change in OXPHOS and MTHFD2 flux directionality with increasing rotenone concentrations could be due to underlying changes in mitochondrial physiology, mitophagy or mitochondrial signalling, which need to be explored in future studies43,44.

Fig. 4: Dynamic flux analysis with parallel tracers to quantify oxidative MTHFD2 flux.
figure 4

a, Oxidative MTHFD2 flux and percentage decrease in OCR following MTHFD2 inhibition in SKOV3 cells treated with rotenone (0, 31, 62, 125, 250, 500 or 1,000 nM) and MTHFD2 inhibitor (0 or 60 µM). Oxidative MTHFD2 flux is proportional to [2H1]-serine/[2H1]-NADH enrichment in cells cultured with 4-[2H]-glucose for 8 h. Percentage decrease in OCR is estimated in SKOV3 cells treated with 60 µM MTHFD2 inhibitor compared with 0 µM MTHFD2 inhibitor. Oxidative flux data are combined from two independent experiments (n = 4), with mean ± s.d. estimated using error propagation. OCR data are representative of two independent experiments and presented as mean ± s.d. estimated using error propagation (n = 5). b, Reductive MTHFD2 flux and basal OCR in SKOV3 cells treated with rotenone (0, 31, 62, 125, 250 or 1,000 nM) and MTHFD2 inhibitor (0 or 60 µM). Reductive MTHFD2 flux is proportional to [2H1]-NADH in cells cultured with 2,3,3-[2H]-serine for 8 h. OCR data are representative of two independent experiments and presented as mean ± s.e.m. (n = 5). c, Schematic of combination tracer experiment with 4-[2H]-glucose and [2H]-formate to track oxidative MTHFD2 flux. Inset: detailed view of [2H] transfer from 4-[2H]-glucose and [2H]-formate to MTHFD2 reaction intermediates, uniquely forming [2H2]-serine via oxidative MTHFD2. d, Fractional labelling of [2H2]-serine in SKOV3 and OVCAR8 cells at 0.5, 1 and 2 h following parallel tracer introduction of 4-[2H]-glucose and [2H]-formate. Data combined from two independent experiments, presented as mean ± s.e.m. (n = 6) and analysed using two-tailed Welch’s t-test. e, Ratio of [2H2]-serine/[2H]-NADH enrichment in SKOV3 and OVCAR8 cells at 2 h after combined tracer introduction of 4-[2H]-glucose and [2H]-formate, representing the normalized contribution of [2H]-NADH to [2H2]-serine via oxidative MTHFD2 flux. Data combined from two independent experiments (n = 6), with mean ± s.d. estimated using error propagation and analysed by two-tailed Welch’s t-test. f, [2H2]-serine enrichment in SKOV3 cells expressing shScramble or shUQCR11 at 2 h after combined tracer introduction of 4-[2H]-glucose and [2H]-formate. Enrichments relative to mean enrichment of [2H2]-serine in SKOV3 shScramble. Data combined from two independent experiments, presented as mean ± s.e.m. (n = 5) and analysed using one-tailed non-parametric Mann–Whitney test.

Source data

Next we employed a combination tracer experiment with 4-[2H]-glucose and [2H]-formate to further deconvolute oxidative mitochondrial MTHFD2 activity (Fig. 4c and Supplementary Information 4). Early time-course measurements ensured that serine enrichment from background [2H] labelling was minimized and remained specific to the oxidative flux of mitochondrial MTHFD2. We observed that enrichment of [2H2]-serine monotonically increased in OVCAR8 cells in the first 120 min of tracer introduction, and was significantly higher at 120 min than in SKOV3 cells in which enrichment remained constant (Fig. 4d). Remarkably, the normalized contribution of [2H] from [2H]-NADH to [2H2]-serine, representing relative oxidative MTHFD2 flux, was significantly higher in OVCAR8 cells compared with SKOV3 cells (Fig. 4e). There was also significantly higher enrichment of serine in isogenic SKOV3 cells expressing shUQCR11, demonstrating the emergence of oxidative MTHFD2 flux when ETC is impaired (Fig. 4f).

We performed global metabolomics profiling to investigate systemic metabolic changes when MTHFD2 was knocked down in UQCR11-null and -intact cells (Fig. 5a). We observed differential levels of metabolites across central carbon, amino acid, purine and pyrimidine metabolism in both UQCR11-null (OVCAR8) and -intact (SKOV3) cells. However, changes in metabolites involved in serine–glycine one-carbon metabolism encompassing MTHFD2 reactions, the methionine cycle, the TCA cycle, OXPHOS and NAD+ metabolism were more pronounced in OVCAR8 cells compared with SKOV3 cells following MTHFD2 knockdown (Fig. 5a). This highlighted the selective reliance of UQCR11-null cells on MTHFD2 and one-carbon metabolic pathways. Further, metabolite set enrichment analysis elucidated differences in the same pathways that were more pronounced in UQCR11-null cells compared with -intact cells following MTHFD2 knockdown (Fig. 5b). Notably, serine, glycine and methionine accumulated in OVCAR8 following MTHFD2 knockdown, signalling a reduction in serine oxidation and a disruption in the coupled folate and methionine cycles, whereas levels of these metabolites were largely unaffected by MTHFD2 knockdown in SKOV3 cells (Fig. 5c). Furthermore, in OVCAR8 cells levels of TCA intermediates citrate, α-ketoglutarate and succinate were reduced whereas fumarate, malate and aspartate were increased following MTHFD2 knockdown (Fig. 5c). These data indicated that TCA cycle reactions upstream of succinate dehydrogenase (complex II) and complex III in the ETC that rely on NAD+/NADH as cofactors were affected due to imbalance of NAD+/NADH within the mitochondria when MTHFD2 was targeted in OVCAR8 cells but not in SKOV3 cells (Fig. 5c). Additionally, MTHFD2 knockdown had no effect on glycolytic intermediates in either OVCAR8 or SKOV3 cells (Fig. 5c), highlighting its targeted influence on mitochondrial metabolism of UQCR11-null cells. Metabolomics profiles of UQCR11-null and -intact ovarian cancer lines from the CCLE panel also show distinction in serine–glycine one-carbon metabolism and its related methionine cycle pathways45 (Extended Data Fig. 8a). Previous reports demonstrated that complex III impairment impedes pyrimidine synthesis and shifts its reliance onto uridine and pyruvate, which warranted investigation of nucleotide profiles of UQCR11-null and -intact cells46,47. We profiled a panel of >100 nucleotides and nucleotide intermediates involved in de novo synthesis, salvage and degradation pathways in parental ovarian cancer lines (Extended Data Fig. 8b–d). Enrichment analysis revealed that synthesis of purine, rather than pyrimidine, in UQCR11-null cells was markedly different compared with that in UQCR11-intact cells (Extended Data Fig. 8b); in fact, out of ten statistically significant (adjusted P < 0.05) and biologically relevant (fold-change >1.5 or <1/1.5), seven are purine intermediates but only two are pyrimidine intermediates (Extended Data Fig. 8c). Remarkably, nucleotide profiling showed that MTHFD2 inhibition affected only a small subset of nucleotides in isogenic SKOV3 models (Extended Data Fig. 8e, red box), with the most marked changes in SKOV3 shScramble cells (ten out 49 differentially abundant metabolites) compared with SKOV3 shUQCR11 (three out of 49) (Extended Data Fig. 8f). K-means clustering of metabolite levels measured in these four groups separated into four clusters revealed a distinction between SKOV3 shScramble cells treated with or without MTHFD2 inhibitor; however, SKOV3 shUQCR11 cells fall within the same cluster regardless of MTHFD2 inhibitor treatment (Extended Data Fig. 8g). Notably, these data support that pyrimidine synthesis is sustained during ETC disruption48 and that these cells maintain pyrimidine levels even when UQCR11 is deficient. This is presumably due to partial ETC impairment, with UQCR11 activity being sufficient to support dihydro-orotate dehydrogenase activity. Notably, de novo pyrimidine synthesis flux measured using [13C4]-aspartate tracer was similarly reduced in SKOV3 and OVCAR8 cells following MTHFD2 inhibition (Extended Data Fig. 8h). Analysis of the gene expression of distinct pyrimidine synthesis pathways in TCGA ovarian tumours also showed no downregulation in UQCR11-null tumours compared with UQCR11-intact (Supplementary Fig. 5). MTHFD2 inhibitor suppressed proliferation of both UQCR11-null parental and SKOV3 shUQCR11 cells compared with UQCR11-intact cells; however, uridine was unable to rescue cells from MTHFD2 inhibition (Extended Data Fig. 9a,b and Supplementary Fig. 6). Similarly, pyruvate could not rescue UQCR11-null cells when MTHFD2 was knocked down in SKOV3 and OVCAR8 cells (Extended Data Fig. 9c and Supplementary Fig. 7). The limited extent of UQCR11 loss in 19p13.3-deleted ovarian tumours did not induce a complex III-dependent reduction in de novo pyrimidine synthesis. Further, MTHFD2 inhibition had a widespread effect on nucleotide metabolism in UQCR11-intact cells, probably due to reliance on reductive MTHFD2. However, its direct effect on pyrimidine synthesis was not exacerbated in UQCR11-null cells. Cumulatively, this suggested that inhibition of MTHFD2 had disrupted serine–glycine one-carbon metabolism as seen in Fig. 4, consequently affecting purine synthesis.

Fig. 5: Metabolic profiling and pathway analysis reveal metabolic compensation and systemic metabolic changes in UQCR11-intact and -null ovarian cancers, respectively, following inhibition of MTHFD2.
figure 5

a, Raindrop plot representing fold-change (FC) of individual metabolites present in each of the metabolic pathways as defined by the KEGG knowledge base. Circle colours represent log(FC) of metabolites in cells expressing shMTHFD2 with respect to shScramble in cell lines OVCAR8 (red) and SKOV3 (blue). Absolute log(FC) values are curtailed at 2.0 to improve visualization. Statistical analysis performed using two-sample t-tests corrected for multiple hypothesis testing, built into MetaboAnalyst 5.0. b, Pathway enrichment analysis based on human metabolic pathways in the KEGG database quantifies metabolic changes following dox-induced expression of shMTHFD2 or shScramble in OVCAR8 and SKOV3 cell lines. Pathway impact and results of statistical tests using MetaboAnalyst 5.0 are reported from n = 5 biological replicates for each condition. c, Differences in metabolites analysed by GC–MS in dox-inducible ovarian cancer lines SKOV3 and OVCAR8 expressing shScramble or shMTHFD2 following treatment with dox (n = 5). Violin plots represent data spread, and dashed and dotted lines represent median and quartiles, respectively. Data analysed using Student’s two-tailed t-test, corrected for multiple hypothesis testing with the Benjamini–Hochberg method. tRNA, transfer RNA; I.S., internal standard.

Source data

Together, dynamic tracing and metabolomics data provided evidence that the collateral lethal gene pairing of UQCR11 and MTHFD2 maintained metabolic compensation via the intricate rewiring of compartmentalized mitochondrial NAD+-producing fluxes. We developed isogenic SKOV3 and OVCAR8 cell lines with NNT, IDH2 or IDH3A knocked down to investigate whether reversible TCA cycle enzymes can act as paralogous NAD+ sources49 (Extended Data Fig. 9d). Importantly, knocking down these genes did not reduce SKOV3 viability (Extended Data Fig. 9e). Interestingly, in UQCR11-null cells, knocking down NNT or IDH3A, both of which can be reversed to produce NAD+, led to a modest 15% reduction in viability compared with the 50% reduction observed with MTHFD2 knockdown (Fig. 2e, Extended Data Fig. 9e and Supplementary Fig. 8). To understand whether these reactions could also supply NAD+ in UQCR11-null cells, we investigated whether their suppression would synergize with MTHFD2 inhibition. Remarkably, the effect of MTHFD2 inhibition was not influenced by these knockdowns, in either UQCR11-intact or -null cells (Extended Data Fig. 9f and Supplementary Fig. 8). These empirical observations agreed with the MOMFA algorithm, which predicted oxidative MTHFD2 as a primary collateral lethal target but not IDH or NNT (Extended Data Fig. 3d). This highlighted the unique ability of CLIM to predict collateral lethal metabolic target pairs that have compensatory metabolic links elucidated via metabolic flux analysis and validated by empirical stable-isotope tracing.

MTHFD2 was predicted and validated as a collateral lethal target in ovarian tumours with 19p13.3 deletion (Figs. 1 and 2). However, distinct genetic backgrounds observed in ovarian tumours can potentially influence their systemic metabolic reprogramming. We looked at those NNMF-stratified tumours (Fig. 1d) with distinct genetic backgrounds to discern whether mutations co-occurring with 19p13.3 loss could confound the collateral lethal effect of targeting MTHFD2. We selected clusters 3, 11, 12 and 32, which had the 19p13.3 deletion as the only overlapping molecular feature, and 28, 5, 24 and 4 unique molecular features, respectively (Fig. 6a). KMT2C and BRCA2 mutations were unique to clusters 3 and 12, respectively, but TP53 mutation and 11p15 deletion were common to both (Fig. 6a,b). Deletions in 1p21 and 22q13 loci were the distinguishing features for clusters 11 and 32, respectively, along with underlying 19q deletion (Fig. 6a,b). Although the dimensionally reduced projections of complete transcriptional profiles were indistinguishable, the metabolic transcriptional profile showed differences between clusters 11 and 32, and their distinction from clusters 3 and 12, which were indistinguishable from each other (Fig. 6c and Extended Data Fig. 10a). Notably, the reconstructed GSMMs for the four clusters also displayed different reactions akin to their metabolic transcriptional profiles (Fig. 6d). Importantly, most of the reactions (413 out of 620) were common across all reconstructed models for the four clusters and the average ovarian cancer model, suggesting the existence of a core metabolic subnetwork unaffected by genetic heterogeneity (Fig. 6d and Extended Data Fig. 10b). We investigated the transcriptomic dissimilarities of these clusters and their influence on the utility of MTHFD2 as a collateral lethal target at a phenotypic level. Comparing the dependency score of MTHFD2 knockdown in ovarian cancer cell lines, as evaluated by the Cancer Dependency Map RNA interference screens, revealed that the correlation between MTHFD2 essentiality and MTHFD2-to-UQCR11 expression difference trended negatively (Fig. 6e). This indicated that higher MTHFD2 and lower UQCR11 expression predicted a higher dependence on MTHFD2 in these lines. Remarkably, CLIM identified that MTHFD2 was always in the top-ranking collateral lethal targets in all four clusters, as evident from the high therapeutic window index (TWI) and Pareto area under the curve (PAUC) in UQCR11-null cells (Fig. 6f and Extended Data Fig. 2c–e). Thus, despite deviations in the metabolic models of UQCR11-null ovarian cancers that have distinct genetic backgrounds, the importance of MTHFD2 as collateral lethal target was maintained.

Fig. 6: MTHFD2 is a potential collateral lethal target across varied genetic backgrounds and microenvironment pressures.
figure 6

a, UpSet plot of distinguishing and overlapping molecular features (CNA and mutations) of the four ovarian tumour clusters with 19p13.3 deletion identified by NNMF in Fig. 1d. Rows in the overlap matrix represent all combinations of overlaps among four clusters; vertical bar chart represents number of ovarian samples in each cluster; horizontal bar chart represents number of molecular features specific to each row’s overlapping cluster combination; dot plot shows cluster-specific scores of molecular features calculated by NNMF in Fig. 1d. b, Dendrogram estimated from the similarity of molecular features of the four HGSOC clusters with 19p13.3 deletion. c, Multidimensional scaling-based projection of metabolic transcriptional profiles of four HGSOC clusters with 19p13.3 deletion. d, Venn diagram displaying overlap of reactions in reconstructed metabolic models illustrating the distinction between four HGSOC clusters with 19p13.3 deletion. e, Correlation of mRNA expression difference (MTHFD2 versus UQCR11) and dependency scores of MTHFD2 in ovarian cancer lines (Cancer DepMap), estimated using Spearman’s rank correlation test (n = 37). f, TWI and PAUC scores of top-ranking collateral lethal candidate targets in reconstructed metabolic models of clusters 3, 12, 11 and 32. g, Single-cell gene expression of MTHFD2 and UQCR11 from scRNA-seq data from ascitic fluid of patients with ovarian cancer, coloured by cell type (top), average expression of one-carbon metabolic signature no. 1 (middle) and serine, glycine and threonine metabolic signature no. 1 (bottom). Single-cell expression data were normalized using MAGIC and used to estimate Spearman’s correlation coefficient. h, CIBERSORTx-derived, cell-type-specific scores projected on two-dimensional (2D) PHATE embedding of stromal scores representing composition of ovarian tumour samples from TCGA. Joy plots reflect the distribution of cell-specific scores across the quadrants of 2D PHATE embedding. i, Projection of 19p13.3 deletion (log2(CNA < –0.4)) on the 2D PHATE plane representing the stromal composition of ovarian tumour samples from TCGA. j, Average expression of genes in one-carbon metabolic signature no. 1 across the quadrants of stromal 2D PHATE embedding. k, mRNA expression of MTHFD2 and UQCR11 across the quadrants of the stromal 2D PHATE embedding.

Source data

Although a varied genetic background can intrinsically influence ovarian cancer metabolism, so too can stromal-rich microenvironments50. The collateral lethal targeting of MTHFD2 will be ineffective if the stroma compensates for metabolic deficiencies in the cancer. We first analysed single-cell RNA sequencing (scRNA-seq) data from ascitic fluid of patients with ovarian cancer and observed that the expressions of MTHFD2 and UQCR11 were strongly negatively correlated across all cell types in ovarian tumours (Fig. 6g). Epithelial (malignant) cells were at the end of the spectrum, with the lowest UQCR11 and highest MTHFD2 expression (Fig. 6g and Extended Data Fig. 10c,d). The pathway-level expression of one-carbon metabolism, including MTHFD2 and encompassing pathways, would reveal their importance in stromal cells when UQCR11 is lost. We first derived ten curated gene sets that would represent pathways related to one-carbon metabolism and ETC from bulk RNA-seq data from the TCGA ovarian tumour cohort (Supplementary Information 5, Supplementary Fig. 9 and Supplementary Table 4). Remarkably, only three of these gene sets were differentially expressed in 19p13.3-deleted ovarian tumours compared with those without deletion from the TCGA cohort (Extended Data Fig. 10e–g). Notably, gene signatures representing the one-carbon pathway including MTHFD2 and serine–glycine–threonine metabolism were both upregulated in 19p13.3-deleted tumours (Extended Data Fig. 10e,f). In contrast, the gene signature reflecting NAD+ biosynthesis was downregulated in 19p13.3-deleted tumours (Extended Data Fig. 10g). This corroborated the predictions from CLIM, highlighting the upregulation of MTHFD2 and related pathways in UQCR11-null tumours. Importantly, at the single-cell level the pathway expression of one-carbon and serine–glycine–threonine metabolism gene signatures was higher in epithelial cell types compared with fibroblasts, lymphocytes and macrophages (Fig. 6g). This highlighted that cancer cells rely on cell-intrinsic MTHFD2 and related pathways to restore NAD+ rather than on one-carbon metabolism of stromal cells, especially when they display UQCR11 loss.

These observations were corroborated from bulk RNA-seq data after deconvoluting malignant and stromal populations using CIBERSORTx. Tumours from the TCGA ovarian cancer dataset displayed the presence of five major populations after deconvolution (Fig. 6h). A dimensionally reduced projection of stromal scores for the five populations was obtained using PHATE to visualize the dominance of different stromal types across the TCGA cohort (Fig. 6h). The first PHATE component distinguished tumours based on dominance of epithelial cells (negative PHATE 1 embedding) or fibroblasts (positive PHATE 1 embedding), while the the second component dissected the dominance of T cells (negative PHATE 2 embedding) and B cells (positive PHATE 2 embedding). Thus, dividing the PHATE plane into four quadrants allowed us to compare four subgroups of tumour with similar numbers of samples across the groups, each with a distinct stromal population (Fig. 6i). The dominant stromal population of each quadrant was discernible by their respective cell-type-specific score distribution (Fig. 6h, joyplots). Notably, the occurrence of 19p13.3 deletion was indistinguishable across both PHATE dimensions (Fig. 6i), indicating that chromosomal alteration was not associated with stromal composition. Remarkably, pathway expressions of the one-carbon and serine–glycine–threonine metabolism signatures had no discernible differences across the quadrants (Fig. 6j and Extended Data Fig. 10h). Further, expression of MTHFD2 was similar across all four quadrants (Fig. 6k). These data indicated that the prevalence of UQCR11-null tumours, and the transcriptional activity of the collateral lethal one-carbon metabolism pathway and MTHFD2, were invariant across ovarian tumours with distinct stromal-dominant populations. Notably, expression of UQCR11 in quadrant Q2 was skewed towards the lower end, in line with the dominance of epithelial cells (Fig. 6h,k). These observations were corroborated by microdissection of an ovarian tumour, where malignant tumour segments showing lower UQCR11 expression compared with paired stromal segments also displayed significantly higher one-carbon metabolism pathway expression (Extended Data Fig. 10i). In contrast, tumours where UQCR11 expression was similar across malignant and stromal tumour segments displayed no difference in one-carbon metabolism pathway expression (Extended Data Fig. 10i). Cumulatively, these observations suggest that MTHFD2 is of importance as a collateral lethal metabolic target in UQCR11-null ovarian cancers, regardless of differences in their intrinsic genetic make-up and extrinsic microenvironments.

To strengthen the validity of collateral lethal targets predicted by CLIM through in silico and in vitro modalities, we developed subcutaneously implanted tumour models representing UQCR11-null and -intact ovarian tumours with dox-inducible knockdown of MTHFD2 (Fig. 7a,b). As per CLIM’s prediction of MTHFD2 essentiality in UQCR11-null cancers, dox-induced knockdown of MTHFD2 dramatically decreased xenograft tumour growth in UQCR11-null but not in UQCR11-intact cells (Fig. 7c,d), despite marked reduction in MTHFD2 expression in UQCR11-intact tumours (Fig. 7e). In line with tumour growth, apoptotic cell populations significantly increased in tissue sections from UQCR11-null tumours (Fig. 7f,g). Collectively, these data reveal the potential of our systems-biology-based workflow to predict collateral lethal metabolic targets in ovarian cancer that show a marked advantage in eliminating tumour growth in vivo.

Fig. 7: Remission of UQCR11-null ovarian tumours on collateral lethal targeting of MTHFD2 in vivo.
figure 7

a,b. Schematic for evaluation of in vivo efficacy of collateral lethal targeting of MTHFD2 in mice implanted with subcutaneous tumours from shMTHFD2-expressing OVCAR8 cells (a) or shMTHFD2-expressing SKOV3 cells (b). c,d, Characterization of tumour size and weight from mice subcutaneously implanted with shMTFHD2-expressing OVCAR8 cells (c) or SKOV3 cells (d) and fed either regular food (ctrl) or food + dox (dox). Data presented as mean ± s.d. for n = 6 mice in each group, and analysed using unpaired two-tailed t-test. e, Validation of dox-induced knockdown on MTFHD2 via immunoblots to measure MTHFD2 protein expression in shMTHFD2-expressing SKOV3-implanted tumours from control and treatment arms. Data representative of two independent experiments. f,g, Representative immunohistochemistry images (f) and quantification (g) of apoptosis marker, cleaved Caspase 3, in tumour sections from mice implanted with shMTHFD2-expressing OVCAR8 cells and fed with ctrl or dox; n = 33 imaged sections for ctrl and n = 20 for dox. Data presented as mean ± s.d. and analysed using two-sided t-test with Welch’s correction.

Source data

Discussion

The utilization of precision medicine-based approaches to discover metabolic targets in cancers has been severely lacking, with families of drugs targeting a given mutation showing drastically differing responses across patients. We therefore used a metabolic systems biology approach to integrate genomic and transcriptomic data with machine learning, genome-scale metabolic flux analysis and state-of-the-art tracing studies to mechanistically identify collateral lethal metabolic vulnerabilities in cancers with specific genomic deletions. The discovery of precision metabolic targets hinges on uncovering redundant metabolic pathways that confer adaptability and survival advantage in cancer cells51,52,53. Importantly, CLIM predicted collateral lethal metabolic pathways and mechanistically explained the compensation between predicted paralogous pathways. Herein we demonstrate the emergence of MTHFD2 as a collateral lethal target in hypomorphic ovarian cancers with UQCR11 copy loss. MTHFD2 has long been considered a potential metabolic target in many cancers due to its overexpression in cancer cells compared with healthy adult cells28,54,55, coupling to folate metabolism-mediated functions39, redox balance56,57 and nucleotide metabolism28,58. Despite a broad empirical knowledge base, the successful use of MTHFD2 inhibitors in the clinic has seen limited progress in ovarian cancers. This study has reinvigorated exploration of the function of MTHFD2 in the context of tumour suppressor gene deletions, especially in cancers with UQCR11 loss. Interestingly, one recent study found that high expression of most complex III subunits predicted higher mortality59. In contrast, our survival analysis showed that tumours with 19p13.3 loss correspond to remarkably higher mortality. Notably, the underlying complex III regulation observed in this study may not be because of large-scale chromosomal alterations, as is the case for UQCR11 loss seen herein. CLIM uncovered an unprecedented non-canonical compensatory function of the MTHFD2 pathway via the oxidative mitochondrial NAD+ balance that is selectively targetable in UQCR11-null ovarian and endometrial cancers. It is important to note that, even with parallel and combinatorial stable-isotope tracing providing a quantitative reflection of oxidative and reductive MTHFD2 fluxes, it is difficult to completely dissect bidirectionality in highly reversible reactions and may require techniques such as continuous in vivo monitoring of metabolism by nuclear magnetic resonance60 to evaluate oxidative MTHFD2 and related pathways in real time. In the future, dynamic modelling of metabolism could improve prediction of collateral lethal metabolic mechanisms in genome-scale models. The modularity of CLIM allows the discovery of mechanistically linked collateral lethal metabolic gene pairs that arise from other chromosomal alterations such as amplifications. Ultimately, CLIM is an end-to-end precision medicine platform that mines clinically relevant chromosomal alterations leading to metabolic vulnerability and predicts collateral lethal metabolic targets linked through mutual metabolic function, thus providing a basis for both empirical and preclinical validation.

Methods

Analysis of clinical and molecular data for tumour samples

Data access

Molecular and clinical data were obtained via CBioPortal61,62 for TCGA Pan-Cancer Atlas samples, CCLE (Broad 2019), NCI-60 Cell Lines and AACR Project GENIE63 (cohort v.7.0-public) samples for HGSOC, UCEC and UCS.

Data processing

For all bioinformatics and machine learning analyses, only primary tumour samples were retained. Gene expression data were downloaded as z-scores relative to all samples for TCGA datasets. Segmentation files were used as input for the GISTIC2.0 algorithm14 deployed on GenePattern Cloud64 with default settings, to obtain linearized CNA values and regions of statistically significant focal deletions.

Metabolic genes

A reference list of metabolic genes for all analyses was obtained from the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. The dataset of metabolic pathways and gene sets were manually curated to remove pathways involved in xenobiotic metabolism, proteasomal processing and signalling.

Genetic paralogues

Paralogous genes for the set of deleted metabolic genes were obtained from the Ensembl database (http://uswest.ensembl.org/Help/View?id=137), determined by the Gene Ontology/Paralogy prediction method. Only those metabolic genes with at least one predicted paralogue in the database were counted as genes with paralogues.

Circos plots

Circos plots were generated using circos-0.69-9 software65. Average copy number gain and loss were used to populate the outermost rings; the middle ring was populated with the GISTIC2.0 –log10(residual q-value) to represent focal deletions of statistical significance; the innermost rings were populated by the number of genes and metabolic genes lying within the peaks of focal deletion detected by GISTIC2.0. The labels identify deleted metabolic genes with their official Human Gene Nomenclature Committee symbol.

Correlation

Metabolic genes deemed to be lost due to chromosomal deletions were selected based on their frequency of loss, as determined by GISTIC2.0 and whether copy loss translated to loss of gene expression. This condition is a representation of loss of metabolic function and is estimated by Spearman’s rank correlation between the linearized CNA and z-score of mRNA expression.

Survival analysis

Cohorts were divided according to CNA or gene expression, as described. Plots were generated and log-rank statistical tests were performed using the function MatSurv (http://github.com/aebergl/MatSurv)66.

Venn diagrams

Diagrams for pictorial representation of overlaps across the reconstructed ovarian cancer metabolic models were generated using the open-source Venn diagram web-tool http://bioinformatics.psb.ugent.be/webtools/Venn/.

Cancer dependency map scores

Genetic dependency data (DEMETER2 scores) for MTHD2 in cell lines were downloaded from the DepMap portal (CRISPR Public 20Q1).

RSF analysis in TCGA ovarian tumours

Ovarian tumour samples from the TCGA Pan-Cancer dataset with both gene expression and clinical data available were analysed (n = 295). RSF models use the machine learning concept of forest ensembles learners for application to right-censored survival data. Here we applied RSF to discover risk factors—in particular, metabolic genes that predict the prognosis of patients with 19p13.3-deleted ovarian tumours. RSF was implemented in R (CRAN 4.0.3) using the package randomForestSRC (cran.r-project.org/web/packages/randomForestSRC). In addition to patient age and expression of enzyme-encoding genes, the following KEGG pathways were used as risk factors for input to the RSF model: one-carbon pool by folate (hsa00670), cysteine and methionine metabolism (has 00270), serine, glycine and threonine metabolism (has00260), ECR reaction (hsa00190), TCA cycle (hsa00020) and NAD pathway (hsa00760). Further justification is provided in Supplementary Methods. Variable importance and mortality (overall survival) prediction plots were generated using the function vimp. Heatmap and variable importance plots are shown for importance values >0.001. Inputs and estimates from the RSF models are shown in Supplementary Table 1.

NNMF for classification of molecular features and patient strata

We used a recursive NNMF approach on integrated mutation and copy number data from 629 patients with ovarian cancer to cluster those with distinct molecular traits—specifically, deep deletions with metabolic genes. Unsupervised NNMF was performed on a molecular feature-by-sample matrix using a method adapted from Moffitt et al.67 and scripted in MATLAB (Mathworks, 2018a). Molecular features were defined by copy number alteration values of focal loci and frequently occurring mutations. Mutation data were represented as binary, where 0 represents no mutation and a positive 'score' represents all but non-sense mutations. The score was set to the 0.5th percentile of linearized copy number values, to ensure that mutations would be ranked as importantly as deletions by the unsupervised algorithm. To ensure a non-negative matrix and to prioritize focal deletions, the combined data were transformed as 2(–x), where x represents every matrix element. For the ovarian cancer dataset, the top 20 frequently occurring mutations as reported by the database COSMIC v.91 (ref. 68) (cancer.sanger.ac.uk) were retained in the feature matrix. Detailed methodology is provided in Supplementary Methods.

UpSet plots

UpSet plots were used to visualize overlap of molecular features from the feature loading in four clusters where 19p13.3 deletion was identified as a defining feature identified by NMF-based clustering of ovarian tumour samples from TCGA. The plots were generated with the UpSetR69 package implemented in R (CRAN 3.6.3).

Multi-objective flux analysis to identify collateral lethal metabolic targets

Multi-objective metabolic flux analysis

We then employed multi-objective flux balance analysis to compute metabolic activity that represents the adaptable nature of cancer cell metabolism. We used Pareto optimization to compute metabolic fluxes under competing metabolic objectives—for example, cellular growth (that is, biomass synthesis) and cell survival (that is, ATP and redox maintenance)70,71. These metabolic objectives are critical in tumorigenesis due to high bioenergetic demands in cancer cells competing for the limited nutrients available in the tumour microenvironment. The design of the multi-objective metabolic flux analysis has been developed in our laboratory and demonstrated in a hepatocyte system70,71. The flux balance analysis algorithm implemented in the toolbox COBRA (v.3.0)72 was modified to a multi-objective optimization problem. We employ the normalized normal constraint method derived for engineering problems73, because it is scale independent in the objective function space. This is especially important when the system variables are intracellular fluxes, which can range across multiple orders of magnitude.

Normalized constraint method for generation of Pareto-optimal solutions (two cellular objectives)

To obtain the Pareto frontier for estimation of optimal flux distributions at the two metabolic objectives, known as the anchor points, maximal biomass synthesis is represented by biomass_reaction flux and maximal ATP by maintenance flux, DM_ATP_c flux. The two distinct optimization problems are defined in equation (1), where g1 and g2 are the metabolic objective functions, S is the stochiometric matrix, v are the intracellular fluxes, and the bounds of intracellular fluxes are vlb and vub:

$${\mathrm{maximize}}\;g_1\;{\mathrm{or}}\;g_2$$
(1)

subject to:

$$Sv = 0$$
$$v_{lb} \le v \le v_{ub}$$

The anchor points, or the optimal solutions of the metabolic objectives, are represented by \({{{\boldsymbol{g}}}}_1^ \ast\) and \({{{\boldsymbol{g}}}}_2^ \ast\), and their corresponding optimal intracellular flux distributions are v1* and v2*. Normalization is done with respect to the ranges of objective function values and is represented as \(\bar g\). The utopia point, which is the theoretical optimal solution for both objectives is \({{{\boldsymbol{g}}}}^{{{\boldsymbol{u}}}}\) (equation (2)) and the normalization factors are estimates as in equation (3):

$$g^u = \left[ {g_1^ \ast \;g_2^ \ast } \right]^T$$
(2)
$$l_1 = g_1\left( {v_2^ \ast } \right) - g_1^ \ast$$
(3a)
$$l_2 = g_2\left( {v_1^ \ast } \right) - g_2^ \ast$$
(3b)

The optimization problem and objective function space (equation (4)) used to generate the Pareto frontier are transformed such that the objective function is normalized in terms of gu, l1 and l2:

$$\bar g^T = \left[ {\frac{{g_1\left( v \right) - g_1^ \ast }}{{l_1}}\frac{{g_2\left( v \right) - g_2^ \ast }}{{l_2}}} \right]$$
(4)

The reduction constraint (equation (5)) is derived using orthogonality with the utopia line, N1 that is the vector in the direction of \(\overline {g_1^ \ast }\) and \(\overline {g_2^ \ast }\):

$$\overline {N_1} = \overline {g_2^ \ast } - \overline {g_1^ \ast }$$
(5)

The utopia line is divided into m1 – 1 segments and the reduction constraint vector is divided into equal segments with \(\partial = \frac{1}{{m_1 - 1}}\) normalized increments to obtain the utopia line \(\overline {X_{Pj}}\), defined as the jth segment of the vector connecting the anchor points gk*, corresponding to each Pareto solution, Pj. Each segment is obtained from the weighted fraction, αkj, of the optimal solutions gk*, where j [1, m1] ∩Z (equations (67)):

$$\overline {X_{Pj}} = \mathop {\sum }\limits_{k = 1}^2 \alpha _{kj}\overline {g_k^ \ast }$$
(6a)
$$0 \le \alpha _{kj} \le 1\;{\mathrm{and}}\mathop {\sum }\limits_{k = 1}^2 \alpha _{kj} = 1$$
(6b)

The normal line \(N_1\) is moved across \(X_{Pj}\) by incrementing \(\alpha _{kj}\) (equation (7)) to restrict the feasible region and leading to a new feasible minimum point:

$$\alpha _{kj} = \left\{ {i \ast \partial } \right\}_{i = 0}^{m - 1}$$
(7)

Solving the succession of optimization problems (equation (8)) with the additional normal constraint (equation (9)) will generate a set of Pareto points for corresponding values of each incremental change in feasible regions:

$${\mathrm{minimize}}\left\{ {\overline {g_2\left( v \right)} } \right\}$$
(8)

subject to:

$$N_1.\left( {\overline {g\left( v \right)} - \overline {X_{Pj}} } \right)^T \le 0$$
(9)
$$Sv = 0$$
$$v_{lb} \le v \le v_{ub}$$

The Pareto points obtained are in normalized space and need to be transformed to original coordinates using the inverse mapping relation:

$$g^{{\mathrm{Pareto}}} = \left[ {\left( {\overline {g_1} l_1 + g_1^ \ast } \right)\quad \left( {\overline {g_2} l_2 + g_2^ \ast } \right)} \right]^T$$

The set of Pareto-optimal solutions, \(g^{{\mathrm{Pareto}}}\), are such that increases in one objective function come at the cost of the other. First, Pareto-optimal solutions for the condition with no deletions are simulated using the cancer-specific metabolic model from the previous step.

Metabolic gene deletion is emulated by first limiting all reactions in the model encoded by the deleted gene to 10% of the flux value in the non-deleted condition. To ensure that all reactions are limited, we use the function findRxnsFromMets in COBRA and the gene–protein reaction (GPR) to find reactions that are catalysed by (i) only the gene-encoded enzyme or (ii) a complex that requires the subunit encoded by the deleted gene, represented in the GPR rules as 'and'.

PAUC

The Pareto area-under-the-curve (PAUC) is a quantification of the extent to which each flux changes across the entire Pareto frontier (Extended Data Fig. 2c). The difference between the PAUC values of fluxes is derived by comparing two conditions, in this case Pareto-optimal fluxes in UQCR11-intact cells and UQCR11-null ovarian cancer models. \({\mathrm{PAUC}}_k^C\) is calculated for each condition across all Pareto-optimal solutions, as described in equations (10) and (11):

$${\mathrm{PAUC}}_k^C = \frac{1}{{N_P^C}}\mathop {\sum }\limits_{i = 1}^{N_P^C - 1} \frac{{v_{k,i + 1}^C + v_{k,i}^C}}{2},k \in {\mathrm{intracellular}}\;{\mathrm{fluxes}},C \in \left\{ {{\mathrm{deleted}},{\mathrm{intact}}} \right\}$$
(10)
$$\Delta {\mathrm{PAUC}}_k = {\mathrm{PAUC}}_k^{{\mathrm{DEL}}} - {\mathrm{PAUC}}_k^{{\mathrm{INT}}}$$
(11)

TWI (sensitivity analysis)

The TWI is an estimation of the extent to which a collateral lethal candidate target (vCL), when inhibited, would affect cells with corresponding metabolic deletion compared with cells without deletion (Extended Data Fig. 2d). First, the sensitivity of perturbing (inhibiting) vCL is estimated on each randomly chosen Pareto solution, p, as the change in the objective function value for every incremental inhibition of vCL and \(\delta v_{\mathrm{CL}}\) (equation (12)). All fluxes except \(v_{CL}\) are restricted to within 25% of their original values. The average sensitivity to vCL is calculated as the square-root of the 2-norm of sensitivities for each objective function (equation (13)). The TWI is the difference in average sensitivities to perturbation of vCL between deleted and non-deleted Pareto-optimal solutions (equation (14)):

$$\begin{array}{l}\frac{{{{\Delta }}f_p^{\,C}}}{{{{\Delta }}v_{{\mathrm{CL}}}}} = \mathop {\sum }\limits_{i = 0}^I \frac{{f_p^{\,C}\left( {v_{{\mathrm{CL}}} - (i + 1) \ast \delta v_{{\mathrm{CL}}}} \right) - f_p^C\left( {v_{{\mathrm{CL}}} - (i) \ast \delta v_{{\mathrm{CL}}}} \right)}}{{\delta v_{{\mathrm{CL}}}}}, \\I = {\mathrm{floor}}\left( {0.9 \ast \frac{{v_{{\mathrm{CL}}}}}{{\delta v_{{\mathrm{CL}}}}}} \right)\end{array}$$
(12)
$$s_{{\mathrm{CL}}}^C = \root {2} \of {{\mathop {\sum }\limits_p \frac{{{{\Delta }}f_p^{\,1,C}}}{{{{\Delta }}v_{{\mathrm{CL}}}}} + \mathop {\sum }\limits_p \frac{{{{\Delta }}f_p^{\,2,C}}}{{{{\Delta }}v_{{\mathrm{CL}}}}}}}$$
(13)
$${\mathrm{TWI}} = s_{{\mathrm{CL}}}^{{\mathrm{DEL}}} - s_{{\mathrm{CL}}}^{{\mathrm{INT}}}$$
(14)
$$p \in \{ {\mathrm{randomly}}\;{\mathrm{chosen}}\;{\mathrm{Pareto}}\;{\mathrm{solutions}}\}$$
$$C \in \left\{ {{\mathrm{deleted,intact}}} \right\}$$

Cell culture

CAOV3 were sourced from the American Type Culture Collection. SKOV3, OVCAR8 and HeyA8 were obtained from the MDACC Cell-line Core Facility and cultured in high-glucose DMEM (Corning, no. 10-017-CM) supplemented with 10% fetal bovine serum (Corning, no. 35-010-CV) and penicillin/streptomycin (Hyclone, no. SV30010) at 37 °C in a 5% CO2 incubator. All cell lines were confirmed as having no mycoplasmal contamination using a Mycoplasma Detection kit (Lonza, no. LT07-118).

Tumour implantation and treatment

Eight-week-old female NU/J mice were purchased from Jackson Laboratories and housed under pathogen-free conditions; by subcutaneous flank injection, 1 × 106 dox-inducible shMTHFD2 OVCAR8 or SKOV3 cells in PBS were introduced. Six to nine days after injection of tumour cells, mice were randomly divided into two groups followed by regular food or dox + food. Tumour size was measured every 3 days using a caliper, and tumour volume was calculated using the standard formula 0.5 × L × W2, where L is the longest diameter and W is the shortest diameter. Based on the animal protocol approved by the Institutional Animal Care and Use Committee of Indiana University School of Medicine, any mouse with tumour size ≥1,500 mm3 was euthanized. In this study, all tumours grown in the mouse model at the endpoint were <1,500 mm3. Housing conditions: ambient room temperature 22 ± 2 °C, 12/12 h light/dark cycle (lights on at 7:00 a.m.) and 30–70% relative humidity.

Multilayer machine learning model to predict 19p13.3 loss and utility of collateral lethal targeting of MTHFD2 in endometrial cancer

We used a two-model system to predict the expected MTHFD2 response from tumour mutation data and expression data for ETC genes. All model training and evaluation was performed via the Python toolbox scikit-learn (scikit-learn.org/) and the XGBoost library74. To train the first model that predicts 19p13.3 loss from gene mutation, we used a list of 54 mutations commonly found in UCEC as reported by COSMIC v.91 (ref. 68). We combined mutation data from 1,176 samples from the GENIE database63 and 579 from the UCEC TCGA Pan-Cancer database. We divided the data from the TCGA cohort into a 20% test set for overall model testing and an 80% set for training and testing of the first model. We combined 80% of TCGA mutation data with those from GENIE and split the combined data into an 80% training dataset and a 20% test dataset. We used the chi-squared test and fourfold recursive feature elimination (RFE) to perform feature selection. The chi-squared test was implemented via chi2() and RFE was implemented via RFECV, with the estimator set to scv (linear support vector classifier) and scoring set to accuracy (justification for RFE provided in Supplementary Methods). Training data for three selected mutations (PTEN, TP53 and POLE) were used to train different classification models, with fivefold cross-validation for hyperparameter selection (classification models: KNN, linear SVM, rbf SVM, AdaBoost, XGBoost, decision tree, random forest, logistic regression, naive bayes, LDA). We applied the models to the test set and selected a decision-tree model that had the best performance on the testing and training sets. Next, we used z-scored expression data for 108 ETC genes for the 579 tumour samples and the 19p13.3 prediction from the first model to train the second model. We used the 20% dataset previously set aside for testing and the remainder for training.

OCR measurement

Live-cell OCR via Resipher

A total of 20,000 cells were seeded into flat-bottomed, 96-well treated plates (Falcon, no. 353072) in complete DMEM culture medium and incubated for 5 h to facilitate cell attachment. The medium was then replaced with another containing different rotenone concentrations (31.0, 62.5, 125, 250 and 500 nM) for 1 h, after which medium was replaced by fresh medium containing rotenone and the MTHFD2 inhibitor, DS18561882 (0 or 60 µM), and the Resipher oxygen-sensing lid (Lucid Scientific) was attached. Live OCR was measured for up to 16 h after attaching the lid.

Mito stress test with the Seahorse analyser

Mitochondrial OCR was measured using a Seahorse XFe96 analyser (Agilent Technologies). Cells were seeded in 96-well Seahorse cell culture plates and incubated at 37 °C with 5% CO2 overnight. Cells were exposed to different concentrations of rotenone (31.0, 62.5, 125, 250 and 500 nM) for 12 h, followed by DS18561882 (MedChemExpress, no. HY-130251) for 3 h. The assay was performed according to the manufacturer’s protocol. In brief, medium was replaced with 180 μl of Seahorse XF medium free from serum and sodium bicarbonate. The Seahorse plate was then incubated in a CO2-free incubator at 37 °C for 45 min before being placed in a Seahorse XFe96 analyser. Sequential measurements of OCR were performed by the injection of the following inhibitors—oligomycin, FCCP and rotenone plus antimycin A—through ports A, B and C, respectively, to calculate basal OCR under different stresses. All data were normalized to total cell protein, as measured by bicinchoninic acid assay.

Flux analysis via [2H]-labelled substrates and metabolomics in vitro

Polar metabolite extraction for the analysis of amino acids, NAD+ and NADH was performed as in our previous studies50,75,76,77,78,79. Cells were cultured in six-well plates with either 3-[2H]-glucose (Cambridge Isotope Labs, no. DLM-3557-PK), 4-[2H]-glucose (Cambridge Isotope Labs, no. DLM-9294-PK) or 2,3,3-[2H]-glucose (Cambridge Isotope Labs, no. DLM-1073-PK) and then, after 3, 6 or 24 h, quenched with 800 µl of ice-cold methanol/water (1:1) solution containing 1 µg of norvaline. Cells were scraped while on ice, followed by the addition of 800 µl of high-performance liquid chromatography-grade chloroform to remove lipid content from the sample matrix. Finally, cell extracts were transferred to microcentrifuge tubes and vortexed vigorously for 30 min at 4 °C. The extracted samples were dried in a SpeedVac (ThermoScientific) for 2–4 h without heat and stored at –80 °C. The drying process was continuously monitored and stopped as soon as samples were completely dry, to minimize metabolite degradation. The detailed methodologies for gas chromatography–mass spectrometry (GC–MS), liquid chromatography–quadropole time-of-flight (LC-QTOF), SCIEX 7500 QTRAP liquid chromatography–tandem mass spectrometry (LC-MS/MS) and ThermoScientific Orbitrap Fusion Lumos Tribrid liquid chromatography–high-resolution mass spectroscopy (LC-HRMS) workflows are provided in Supplementary Methods.

scRNA-seq data clustering for gene and pathway score projections

Data access

scRNA-seq data were obtained from an online repository with accession no. GSE146026 (ref. 80). The dataset consists of 11,000 single cells from 22 ovarian cancer ascites samples.

Data processing

Data normalization and visualization were performed using the packages Rmagic81 and PhateR82 implemented in R (CRAN 4.0.4). Raw counts are quantified as log2(TPM/10 + 1), where TPM denotes transcripts per million. We performed an initial quality control that involves removal of genes expressed in fewer than ten cells and exclusion of cells with <1,000 counts. L1 normalization was performed on quality-control-treated data, followed by square-root transformation of the resulting matrix. This was followed by final normalization using the 'magic' function of the package Rmagic81. Briefly, the magic function will first right-multiply the input matrix with a coefficient matrix called Markov affinity matrix and then rescale the matrix. Finally, the various clusters present in the datasets were visualized using the function phate in the package PhateR82 with the following parameters: knn = 5, decay = 100 and t = 20. Clusters were identified based on the following gene markers: epithelial (KRT17, KRT4, EPCAM, MMP7); fibroblasts (PDPN, DCN, THY1, SNAI2); macrophages (CD14, AIF1, CSF1R, CD163); T cells (CD2, GZMB); and B cells (CD19, CD79A). Plots were generated with ggplot2 (ref. 83) implemented in R (CRAN 4.0.4).

Estimation of cell type proportions in ovarian bulk RNA-seq data using CIBERSORTx

Bulk RNA-seq data for ovarian serous cystadenocarcinoma from the TCGA Pan-Cancer Atlas were obtained via CBioportal62,84. scRNA-seq data were obtained from an online repository with accession no. GSE118828 (ref. 85). scRNA-seq was obtained from 14 samples collected from nine patients with varying grades of ovarian cancer. Raw ovarian scRNA-seq data were obtained as unique molecular identifier (UMI) gene counts per cell. Using the pipeline Seurat v.4 (ref. 86) in R (CRAN 4.04), outlier cells with total UMI count <200 or >5,000 were deselected. In addition, dying cells with >5% mitochondrial genes were excluded from further analysis. Quality-control-treated data were log-normalized with a scale factor of 10,000, and the top 2,000 variable features (genes) among cells were identified. Principal component analysis (PCA) was performed on variable features, with the first 20 PCA used for clustering with the shared nearest-neighbour algorithm at a resolution of 0.5 followed by visualization using the uniform manifold approximation and projection dimensional reduction algorithm. Cell type markers are provided in Supplementary Methods. The generated single-cell reference matrix file and bulk RNA-seq were the two main inputs to the online CIBERSORTx digital cytometry87. Cell fractions were computed in absolute mode with 500 permutations.

Reporting summary

Further information on research design is available in the Nature Research Reporting Summary linked to this article.