Abstract
Immune checkpoint inhibitors have improved the clinical management of some cancer cases, yet patients still fail to respond to immunotherapy. Dysregulated metabolism is a common feature of many cancers, and metabolites are known to modulate functions in cancer cells. To identify potential metabolic pathways involved in anti-tumor immune response, we employed a metabolic inhibitor-based drug screen in human lung cancer cell lines and examined expression changes in a panel of immune regulator genes. Notably, pharmacologic inhibition of dihydrofolate reductase (DHFR) downregulated cancer cell expression of cluster of differentiation 24 (CD24), an anti-phagocytic surface protein. Genetic modulation of DHFR resulted in decrease of CD24 expression, whereas tetrahydrofolate, the product of DHFR, enhanced CD24 expression. DHFR inhibition and the consequent CD24 decrease enhanced T cell-mediated tumor cell killing, whereas replenishment of DHFR or CD24 partially mitigated the immune-mediated tumor cell killing that resulted from methotrexate treatment in cancer cells. Moreover, publicly available clinical data analyses further revealed the link between DHFR, CD24, and the antitumor immune response in lung cancer patients. Our study highlights a novel connection between folate metabolism and the anti-tumor immune response and partially interprets how DHFR inhibitors lead to clinical benefits when combined with cancer immunotherapy agents.
Keywords: dihydrofolate reductase (DHFR), CD24, cancer metabolism, immune checkpoints, immune-mediated tumor killing, lung cancer
INTRODUCTION
The advent of immunotherapy has revolutionized the treatment of cancer, and immune checkpoint inhibitors (ICIs) are now used to treat a variety of cancer types (1). However, clinical response rates are below 30% in most solid tumors and many cancer patients are unresponsive to ICI treatments (2). For instance, non-small cell lung carcinoma (NSCLC) patients with KRas/LKB1 co-mutations are known to receive little therapeutic benefit from ICIs (3). Immune checkpoint molecules such as PD-1 and CTLA-4 are key targets in cancer immunotherapy (4). Emerging evidence suggest that cluster of differentiation 24 (CD24), an anti-phagocytic surface protein that signals through macrophage Siglec-10, is a promising target for cancer immunotherapy (5). However, the regulation mechanism of CD24 in human cancers is not fully understood.
Dysregulated cellular metabolism such as the Warburg Effect, is a hallmark of many cancers. In addition to providing materials for bioenergetics and biosynthesis, metabolites also serve as important co-factors to regulate cell signaling and gene expression (6). DHFR is an essential enzyme in de novo pyrimidine synthesis that catalyzes the conversion of dihydrofolate to tetrahydrofolate (THF). DHFR is commonly overexpressed in various cancer types to facilitate DNA replication and tumor proliferation (7). Pharmacological inhibitors of DHFR such as methotrexate and pemetrexed have been used in the clinic for many years to treat various cancers including NSCLC. Methotrexate is FDA-approved for use in acute lymphoblastic leukemia, breast cancer, osteosarcoma, head and neck cancer, advanced non-Hodgkin lymphoma, and lung cancer, while pemetrexed is approved for advanced NSCLC (8,9). Given the crucial metabolic role of DHFR in cancer and use of DHFR pharmacological inhibitors in the clinic, it is important to understand how drugs like methotrexate affect anti-tumor immune escape and patient response to subsequent ICI therapy.
Here, we screen for a novel metabolic enzyme involved in the gene expression of immune checkpoint (IC) regulators in NSCLC and uncover a unique connection between the enzyme DHFR and the innate immune checkpoint regulator CD24 in lung cancer.
RESULTS
Methotrexate decreases CD24 gene expression in lung cancer cells
To gain insight into the link between tumor metabolism and antitumor immunity, we examined the effect of metabolic pathway inhibition on the expression of immune checkpoint regulator genes. We employed a phenotypic drug screen using a customized metabolic inhibitor library targeting thirteen enzymes. These 13 enzymes were selected from a group of 219 enzymes involved in key metabolic pathways implicated in cancer. Furthermore, respective metabolites of the selected enzymes were not reported to regulate gene expression. The screen involved treating the A549 lung cancer cell line with two doses of each inhibitor targeting an individual metabolic enzyme and subsequently measuring gene expression of a panel of immune checkpoint regulators implicated in cancer (Table 1). Inhibition of metabolic enzymes using buthionine sulphoximine (BSO), methotrexate, or lometrexol resulted in significant gene expression decrease of multiple immune checkpoint regulators including PVR, CD24, and IDO1, respectively in a dose-dependent manner (Fig. 1A). Among these changes, decrease of CD24 gene and protein expression by time-dependent pharmacological inhibition of dihydrofolate reductase (DHFR) using methotrexate was further confirmed in A549 and H157 cell lines (Fig. 1B and 1C). These data suggest that DHFR activity is involved in induction of anti-phagocytic CD24 expression in lung cancer cells.
Table 1.
Cancer-related metabolic targets and related inhibitors
| Target enzymes | Genes | Pathway involved | Inhibitor | Ref. |
|---|---|---|---|---|
| Phosphofructokinase 2 | PFKFB3 | Glycolysis | 3-(3-Pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) | (17) |
| Diacylglycerol acyltransferase 1 | DGAT1 | Triglyceride synthesis | A922500 | (29) |
| AICAR transformylase | ADE17 | De novo purine biosynthesis | ATIC dimerization inhibitor | (22) |
| Dihydroorotate dehydrogenase | DHODH | De novo pyrimidine biosynthesis | Brequinar | (19) |
| Glutamate cysteine ligase | GCL | Glutathione biosynthesis | L-Buthionine-(S,R)-Sulfoximine (BSO) | (23) |
| Monoamine oxidase A | MAOA | Tryptophan metabolism | Clorgyline | (24) |
| GTP cyclohydrolase 1 | GCH1 | Non-essential amino acid synthesis | 2,4-Diamino-6-hydroxypyrimidine (DAHP) | (26) |
| GAR transformylase | GART | De novo purine biosynthesis | Lometrexol | (28) |
| Malic enzyme 1 | ME1 | Anaplerosis | Malic enzyme 1 (ME1) inhibitor | (18) |
| Dihydrofolate reductase | DHFR | Thymidine synthesis | Methotrexate | (25) |
| Glucose-6-phosphate dehydrogenase | G6PD | Oxidative PPP | Polydatin | (21) |
| Lanosterol synthase | LSS | Cholesterol biosynthesis | RO 48–8071 fumarate | (20) |
| Delta(24)-sterol reductase | DHCR24 | Cholesterol biosynthesis | Triparanol | (27) |
Figure 1. Metabolic inhibitor screen identifies cancer-related enzymes including DHFR as potential immune gene regulators.

(A) Primary screen examining 12 known immune checkpoint (IC) regulators expression in lung cancer cells treated with inhibitors targeting 13 cancer-related metabolic enzymes. A549 cells were treated with reported working concentrations for 24 hours. Relative mRNA levels of IC regulators were measured via qRT-PCR with GAPDH as a control. (B and C) Effect of targeting DHFR using methotrexate on levels of CD24 gene (B) and CD24 protein (C). A549 (top panels) and H157 (bottom panels) cells were treated with methotrexate at 2 μM for 24 and 48 hours. CD24 mRNA levels and protein levels were determined by qRT-PCR and flow cytometry analysis. Representative FACS plots are shown on the right for (C). Data are mean ± SD from 3 replicates. Two-way ANOVA was used for statistics (ns: not significant; *: 0.01 < p < 0.05; **: p < 0.01).
DHFR and the metabolic product THF induce CD24 expression in cancer cells
To investigate whether DHFR contributes to CD24 gene expression, lung cancer cells were genetically modulated with DHFR and consequent changes in CD24 were monitored. We first assessed the impact of genetically target-downregulating DHFR on CD24 expression in lung cancer cells. Stable knockdown of DHFR significantly decreased CD24 at both gene and protein levels (Fig. 2A). We next determined whether DHFR overexpression could confer CD24 induction in lung cancer cells. We observed that transient overexpression of DHFR in A549 cells significantly enhances CD24 at both the RNA and protein level (Fig. 2B). Furthermore, supplementation with DHFR’s metabolic product, THF, also enhanced CD24 expression in lung cancer cell lines in a dose-dependent manner (Fig. 2C and 2D). These findings indicate that DHFR and its product THF are involved in regulation of CD24 expression in lung cancer.
Figure 2. Genetic modulation of DHFR alters expression of CD24 in lung cancer.

(A and B) Effect of DHFR knockdown (A) or overexpression (B) on CD24 gene expression and protein level. A549 cells were stably transduced with lentiviral DHFR shRNA for knockdown or transiently transfected with pLenti-V5-DHFR for overexpression. CD24 mRNA levels and protein amount on the cell surface were assessed by qRT-PCR and flow cytometry analysis, respectively. Knockdown or overexpression of DHFR was determined by DHFR qRT-PCR. (C and D) Effect of THF treatment on CD24 gene expression in lung cancer cells. A549 (C) and H157 (D) cells were treated with indicated concentrations of THF for 24 hours. CD24 mRNA levels were determined by qRT-PCR and normalized to GAPDH. Data are mean ± SD from 3 replicates. P values were determined by two-tailed unpaired Student’s t test (A-B) and one-way ANOVA (C-D) (ns: not significant; *: 0.01 < p < 0.05; **: p < 0.01).
Targeting DHFR in cancer cells enhances T cell-mediated tumor cell killing in part by signaling through CD24
We next examined whether DHFR is important for immune evasion in lung cancer. Through a co-culture model with lung cancer cells and human peripheral blood mononuclear cells (PBMCs) activated by anti-CD3 and anti-CD28 co-stimulation, we demonstrated that targeting DHFR with methotrexate results in enhanced immune-mediated tumor cell killing and reduced viability of A549 cells when co-cultured with activated T cells (Fig. 3A). In agreement, we observed that the treatment of methotrexate decreases CD24 gene expression in the presence of activated human PBMCs (Fig. 3B).
Figure 3. Methotrexate treatment attenuates CD24 expression and induces immune cell-mediated tumor cell killing.

(A) Effect of targeting DHFR using methotrexate on cancer cell viability in the presence and absence of activated primary human T cells. T cells in human peripheral blood mononuclear cells (PBMC) were activated by CD3/CD28/CD2 and IL2 followed by co-culture with A549 cancer cells for 48 hours treated with 2 μM of methotrexate. Immune cells were washed away, and cancer cell viability was measured by crystal violet assay. (B) CD24 levels were assessed by qRT-PCR in A549 cells co-cultured with primary activated T cells treated with methotrexate as described in (A). (C) Effect of methotrexate on T cell proliferation. Activated and CFSE-stained T cells in human PBMCs were cultured alone (left) or with A549 cancer cells (right) for 72 hours with or without 2 μM of methotrexate. T cell proliferation rate was determined by assessing CFSE stained cells through flow cytometry analysis. (D) Pretreatment of cancer cells with methotrexate enhances T cell-mediated immune cell killing. A549 cells were pretreated with 2 μM of methotrexate for 48 hours. A549 cells were washed and co-cultured with activated T cells as described in (A). Data are mean ± SD from 3 replicates. P values were determined by one-way ANOVA (*: 0.01 < p < 0.05; **: p < 0.01).
To further investigate whether the effect of methotrexate on immune-mediated tumor cell killing is through the inhibition of DHFR, specifically in cancer cells, we first examined the effect of methotrexate on T cell proliferation. Methotrexate enhanced T cell activity not when the drug was applied to T cells cultured alone but when these T cells were co-cultured with cancer cells (Fig. 3C). Moreover, pretreatment of cancer cells with methotrexate in the absence of T cells still enhanced T cell-mediated tumor cell killing (Fig. 3D). These data suggest that DHFR in cancer cells contributes to cancer immune evasion.
We next examined whether the DHFR-CD24 axis is involved in methotrexate-mediated immune-tumor cell killing. Forced expression of DHFR or CD24 significantly attenuated the methotrexate-induced tumor cell killing by T cells (Fig. 4A and 4B). These data suggest the methotrexate effect on T cell-mediated tumor cell killing occurs at least in part by inhibition of DHFR and the downstream effector CD24.
Figure 4. Overexpression of DHFR-CD24 in cancer cells enhances tumor immune evasion.

(A and B) Effect of DHFR (A) or CD24 (B) overexpression on methotrexate-mediated cancer cell killing in the presence of T cells. A549 cells transfected with vectors containing V5 tagged DHFR or CD24 were co-cultured with activated primary human T cells in the presence or absence of 2 μM of methotrexate for 48 hours. Cancer cell viability was assessed by crystal violet assay. The ectopic expression of DHFR and CD24 was analyzed by immunoblotting using anti- V5 antibody. Data are mean ± SD from 3 replicates. P values were determined by one-way ANOVA (ns: not significant; **: p < 0.01).
DHFR, CD24, and immune infiltration are clinically linked in human lung cancer
We next demonstrated the clinical relevance of our finding using publicly available databases. We found that DHFR is associated with poor clinical outcome in lung cancer patients. Overall survival rates were significantly lower in the patient group with high DHFR expression compared to the group with low DHFR expression (Fig. 5A). According to the TIDE database, there exists a significant negative correlation between CD24 mRNA expression and tumor infiltrating cytotoxic T lymphocyte levels in lung cancer patients (Fig. 5B). Furthermore, CD24 mRNA levels were lower in lung adenocarcinoma patients who received therapy containing DHFR inhibitor pemetrexed compared to patients who did not receive DHFR inhibitor-containing therapy (Fig. 5C). These data are in support of our finding that DHFR contributes to CD24 expression which is associated with tumor immune evasion and poor prognosis.
Figure 5. DHFR and CD24 expression negatively correlates with patient outcome and antitumor immunity in patients with lung cancer.

(A) Kaplan-Meier plot of lung cancer patients stratified by DHFR tumor mRNA expression. Highest and lowest 20% of DHFR mRNA expression groups were used to estimate overall survival in TCGA lung adenocarcinoma (LUAD) or lung squamous cell carcinoma (LUSC) patients. DHFR high (n=193) and DHFR low (n=192). (B) Correlation analysis of CD24 mRNA expression z-scores and tumor infiltrating cytotoxic T lymphocyte level z-scores in patients with LUAD or LUSC. Data are obtained from TIDE database patient cohort GSE50081 (n=181). (C) CD24 mRNA expression in LUAD patients treated with non-DHFR inhibitor (n=467) and DHFR inhibitor pemetrexed (n=57). Clinical information and CD24 RSEM were downloaded from Firebrowse. The log-rank test (A), Pearson correlation (B) and Welch two sample t-test (C) were used for statistical analyses.
DISCUSSION
Metabolic reprogramming is a common feature of cancer and has been researched extensively in the context of tumor bioenergetics and biosynthesis. In recent years, dysregulated cellular metabolism has been linked to tumor immune evasion, but the detailed molecular mechanisms through which altered metabolic pathways in cancer cells contribute to immune escape are still not well-understood. Here we seek to better understand the unique connection between lung cancer metabolism and anti-tumor immunity by investigating how the targeting of metabolic enzymes commonly implicated in cancer could influence the expression of genes related to immune regulation. Our findings delineate that cancer cell DHFR, an enzyme important for folate metabolism, is involved in regulating the anti-tumor immune response in part through the regulation of CD24: an innate immune checkpoint protein expressed on breast and ovarian cancer cells which prevents phagocytosis by macrophages.
Our study provides evidence that CD24 regulation mediated by DHFR involves its enzyme activity since pharmacological DHFR inhibition and supplementation of the metabolic product, THF, resulted in decreased and increased expression of CD24, respectively. Metabolites like succinate and alpha-ketoglutarate are reported to affect gene expression through allosteric modulation of epigenetic factors (10). Here we show that the immunomodulatory effects of DHFR in cancer cells is mediated through its metabolic product, THF. Further detailed biochemical and biophysical studies are warranted to elucidate the mechanism by which the enzyme activity and related metabolite THF and the NADP+/NADPH ratio are involved in CD24 regulation process. In addition, a previous report suggests that oncogenic Ras, which is activated in approximately 30% of all cancers, is a suppressor of CD24 mRNA and protein (11). Since oncogenic Ras signaling is prevalent in NSCLC, it would be worthwhile to further investigate how crosstalk between oncogenic Ras signaling and DHFR metabolic activity in lung cancer work to dynamically regulate the expression of the immune factor CD24.
Our clinical correlation studies using publicly available databases demonstrate that DHFR-CD24 expression is associated with poor survival and inversely correlates with the percent of tumor-infiltrating lymphocytes in lung cancer patients (12). DHFR inhibitors such as methotrexate and pemetrexed are widely used in the clinic to treat lung and other types of cancers (13). A recent study suggests that the combination of pembrolizumab and pemetrexed results in significantly higher rates of response and longer progression-free survival in NSCLC patients (14). From a clinical perspective, our finding may provide a clue that explains the therapeutic advantage of the combination, showing that it may be in part due to the CD24 mitigation mediated by DHFR inhibition, which may provide an additive effect with pembrolizumab on enhancing anti-tumor immune response.
EXPERIMENTAL PROCEDURES
Reagents
RO 48–8071 fumarate, clorgyline hydrochloride, malic enzyme inhibitor ME1 were obtained from MedChemExpress. Lometrexol and BSO were from Cayman Chemical. A922500 and brequinar were from Selleck Chemicals. Polydatin was from Santa Cruz Biotechnology. All other metabolic inhibitors used in the study were from Sigma Aldrich. Tetrahydrofolate was obtained from Sigma-Aldrich. Lentiviral short hairpin RNA clone for DHFR was from Dharmacon (sense 5’-CCTGAGAAGAATCGACCTTTA-3’). pLenti6.3/V5-DEST vector encoding DHFR and pLX304-V5-CD24 are obtained from DNASU (HsCD00940668 and HsCD00442245). Primers for HLA-A (forward 5’-GAGGAGGAAGAGCTCAGATAGA-3’ and reverse 5’-GGCAGCTGTCTCACACTTTA-3’) and all other commercially available primers for quantitative RT-PCR were obtained from Integrated DNA Technologies. FITC-conjugated anti-human CD24 antibody (SN3/ab30350) was purchased from Abcam. Anti-V5 antibody (sc-81594) and anti-β-actin antibody (AC-74/A2228) are from Santa Cruz Biotechnology and Sigma-Aldrich, respectively. CellTrace™ CFSE Cell Proliferation Kit (C34554A) is from Life Technologies.
Cell culture and lentiviral infection
A549 and H157 cell lines were obtained from American Type Culture Collection (ATCC) and cultured in RPMI 1640 medium with 10% fetal bovine serum (FBS). 293T cells were cultured in DMEM with 10% FBS. Lentivirus production, stable gene knockdown and overexpression in human cancer cells were performed as described (15,16).
Metabolic inhibitor-based screen
Screening was performed using a customized inhibitor library, which targets metabolic pathways implicated in cancer. 2×105 A549 cells were seeded into 6-well plates and treated with vehicle or two doses of each inhibitor targeting an individual metabolic enzyme. The dosages for each of the 13 inhibitors were determined based on previous literatures (17–29). Expression levels of the 12 immune checkpoint regulator genes were assessed after 24 hours of inhibitor treatment via quantitative RT-PCR.
Quantitative RT-PCR and flow cytometry analysis
The total RNA of A549 and H157 were isolated using RNeasy kit (QIAGEN) followed by reverse transcription of RNA (Applied Biosystems). qRT-PCR was performed using SYBR Green Supermix (Bio-Rad). The CD24 protein level was assessed by flow cytometric staining with anti-human CD24-FITC antibody (BD FACSymphony) and data were analyzed using FlowJo software.
T cell-mediated tumor cell killing assay
Human peripheral blood mononuclear cells (PBMC) were cultured in ImmunoCult-XF T cell expansion medium with ImmunoCult Human CD3/CD28/CD2 T cell activator (STEMCELL Technologies) and 10 ng/mL of IL-2 for one week according to the manufacturer’s protocol. The experiments were performed in RPMI1640 medium with anti-CD3 antibody (500 ng/mL) and IL-2 (10 ng/mL). 2 × 104 A549 cells were plated into a 12-well plate and then co-incubated for 48 hours with activated T cells in the presence or absence of 2 μM of Methotrexate. The cancer cells-to-activated T cells ratio of 1:10 was used. T cells and cell debris were removed and live cancer cells were stained with crystal violet and quantified by a spectrometer at OD 570 nm (30).
T cell proliferation assay
Human primary T cells were activated in T cell expansion medium using ImmunoCult Human CD3/CD28/CD2 T cell activator and 10 ng/mL of IL-2 for a week. To investigate the effect of MTX on T cell proliferation in co-culture or only t-cell culture, T cells were stained with 5 μM of CellTrace™ CFSE dye and maintained in RPMI medium supplemented with human CD3/CD28/CD2 T cell activator and 10 ng/mL of IL-2 for three days. T cell proliferation rates were assessed by analyzing CFSE fluorescence intensity using a flow cytometer.
Bioinformatics and statistical analysis
Gene-expression data and clinical information from TCGA were downloaded from Broad GDAC Firehose. Correlation analysis between z scores of tumor infiltrated cytotoxic T lymphocytes and CD24 expression were obtained from Tumor Immune Dysfunction and Exclusion (TIDE) database (31). Statistical analyses and graphical presentations were performed using GraphPad Prism 9. Data with error bars represent mean ± SD from three replicates. Statistical analysis of significance was based on two-way ANOVA for Figure 1A, two-tailed unpaired Student’s t test for Figures 2A, 2B, and 3C, one-way ANOVA for Figures 3 and 4, log-rank test for Figure 5A, Pearson correlation for Figure 5B, and Welch two sample t-test for Figure 5C.
ACKNOWLEDGEMENTS
This study was supported in part by NIH/NCI grants R01 CA175316 (S.K.), R01 CA207768 (S.K.), R01 CA266613 (S.K.), F99 CA264407 (A.C.B.), P01 CA257906 (H.F and S.R), DoD grant W81XWH-21-1-0213 (S.K.), and the resource of Winship Cancer Institute of Emory University under NIH/NCI P30 CA138292. A.C.B. is an NIH pre-doctoral fellow. S.K. is an American Cancer Society Basic Research Scholar.
Footnotes
CONFLICT OF INTEREST
The authors have no conflict of interest to declare.
DATA AVAILABILITY
All data are contained within the article.
REFERENCES
- 1.Esfahani K, Roudaia L, Buhlaiga N, Del Rincon SV, Papneja N, and Miller WH Jr. (2020) A review of cancer immunotherapy: from the past, to the present, to the future. Curr Oncol 27, S87–S97 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Das S, and Johnson DB (2019) Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer 7, 306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Skoulidis F, Goldberg ME, Greenawalt DM, Hellmann MD, Awad MM, Gainor JF, Schrock AB, Hartmaier RJ, Trabucco SE, Gay L, Ali SM, Elvin JA, Singal G, Ross JS, Fabrizio D, Szabo PM, Chang H, Sasson A, Srinivasan S, Kirov S, Szustakowski J, Vitazka P, Edwards R, Bufill JA, Sharma N, Ou SI, Peled N, Spigel DR, Rizvi H, Aguilar EJ, Carter BW, Erasmus J, Halpenny DF, Plodkowski AJ, Long NM, Nishino M, Denning WL, Galan-Cobo A, Hamdi H, Hirz T, Tong P, Wang J, Rodriguez-Canales J, Villalobos PA, Parra ER, Kalhor N, Sholl LM, Sauter JL, Jungbluth AA, Mino-Kenudson M, Azimi R, Elamin YY, Zhang J, Leonardi GC, Jiang F, Wong KK, Lee JJ, Papadimitrakopoulou VA, Wistuba II, Miller VA, Frampton GM, Wolchok JD, Shaw AT, Janne PA, Stephens PJ, Rudin CM, Geese WJ, Albacker LA, and Heymach JV (2018) STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma. Cancer Discov 8, 822–835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Seidel JA, Otsuka A, and Kabashima K (2018) Anti-PD-1 and Anti-CTLA-4 Therapies in Cancer: Mechanisms of Action, Efficacy, and Limitations. Front Oncol 8, 86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Barkal AA, Brewer RE, Markovic M, Kowarsky M, Barkal SA, Zaro BW, Krishnan V, Hatakeyama J, Dorigo O, Barkal LJ, and Weissman IL (2019) CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature 572, 392–396 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lv H, Lv G, Chen C, Zong Q, Jiang G, Ye D, Cui X, He Y, Xiang W, Han Q, Tang L, Yang W, and Wang H (2021) NAD(+) Metabolism Maintains Inducible PD-L1 Expression to Drive Tumor Immune Evasion. Cell Metab 33, 110–127 [DOI] [PubMed] [Google Scholar]
- 7.Matherly LH, Taub JW, Wong SC, Simpson PM, Ekizian R, Buck S, Williamson M, Amylon M, Pullen J, Camitta B, and Ravindranath Y (1997) Increased frequency of expression of elevated dihydrofolate reductase in T-cell versus B-precursor acute lymphoblastic leukemia in children. Blood 90, 578–589 [PubMed] [Google Scholar]
- 8.Selawry O, Krant M, Scotto J, Kazam E, Schneiderman M, Olson K, Shnider B, Edmonson J, Holland J, and Taylor S 3rd. (1977) Methotrexate compared with placebo in lung cancer. Cancer 40, 4–8 [DOI] [PubMed] [Google Scholar]
- 9.Hanna N, Shepherd FA, Fossella FV, Pereira JR, De Marinis F, von Pawel J, Gatzemeier U, Tsao TC, Pless M, Muller T, Lim HL, Desch C, Szondy K, Gervais R, Shaharyar, Manegold C, Paul S, Paoletti P, Einhorn L, and Bunn PA Jr. (2004) Randomized phase III trial of pemetrexed versus docetaxel in patients with non-small-cell lung cancer previously treated with chemotherapy. J Clin Oncol 22, 1589–1597 [DOI] [PubMed] [Google Scholar]
- 10.Liu PS, Wang H, Li X, Chao T, Teav T, Christen S, Di Conza G, Cheng WC, Chou CH, Vavakova M, Muret C, Debackere K, Mazzone M, Huang HD, Fendt SM, Ivanisevic J, and Ho PC (2017) α-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat Immunol 18, 985–994 [DOI] [PubMed] [Google Scholar]
- 11.Pallegar N, Ayre DC, and Christian S (2015) Repression of CD24 surface protein expression by oncogenic Ras is relieved by inhibition of Raf but not MEK or PI3K. Front Cell Dev Biol 3, 47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zhu CQ, Ding K, Strumpf D, Weir BA, Meyerson M, Pennell N, Thomas RK, Naoki K, Ladd-Acosta C, Liu N, Pintilie M, Der S, Seymour L, Jurisica I, Shepherd FA, and Tsao MS (2010) Prognostic and predictive gene signature for adjuvant chemotherapy in resected non-small-cell lung cancer. J Clin Oncol 28, 4417–4424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Raimondi MV, Randazzo O, La Franca M, Barone G, Vignoni E, Rossi D, and Collina S (2019) DHFR Inhibitors: Reading the Past for Discovering Novel Anticancer Agents. Molecules 24, 1140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, Domine M, Clingan P, Hochmair MJ, Powell SF, Cheng SY, Bischoff HG, Peled N, Grossi F, Jennens RR, Reck M, Hui R, Garon EB, Boyer M, Rubio-Viqueira B, Novello S, Kurata T, Gray JE, Vida J, Wei Z, Yang J, Raftopoulos H, Pietanza MC, Garassino MC, and Investigators, K.-. (2018) Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N Engl J Med 378, 2078–2092 [DOI] [PubMed] [Google Scholar]
- 15.Jin L, Li D, Lee JS, Elf S, Alesi GN, Fan J, Kang HB, Wang D, Fu H, Taunton J, Boggon TJ, Tucker M, Gu TL, Chen ZG, Shin DM, Khuri FR, and Kang S (2013) p90 RSK2 mediates antianoikis signals by both transcription-dependent and -independent mechanisms. Mol Cell Biol 33, 2574–2585 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jin L, Li D, Alesi GN, Fan J, Kang HB, Lu Z, Boggon TJ, Jin P, Yi H, Wright ER, Duong D, Seyfried NT, Egnatchik R, DeBerardinis RJ, Magliocca KR, He C, Arellano ML, Khoury HJ, Shin DM, Khuri FR, and Kang S (2015) Glutamate dehydrogenase 1 signals through antioxidant glutathione peroxidase 1 to regulate redox homeostasis and tumor growth. Cancer cell 27, 257–270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Clem B, Telang S, Clem A, Yalcin A, Meier J, Simmons A, Rasku MA, Arumugam S, Dean WL, Eaton J, Lane A, Trent JO, and Chesney J (2008) Small-molecule inhibition of 6-phosphofructo-2-kinase activity suppresses glycolytic flux and tumor growth. Mol Cancer Ther 7, 110–120 [DOI] [PubMed] [Google Scholar]
- 18.Fernandes LM, Al-Dwairi A, Simmen RCM, Marji M, Brown DM, Jewell SW, and Simmen FA (2018) Malic Enzyme 1 (ME1) is pro-oncogenic in Apc(Min/+) mice. Sci Rep 8, 14268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.He T, Haapa-Paananen S, Kaminskyy VO, Kohonen P, Fey V, Zhivotovsky B, Kallioniemi O, and Perala M (2014) Inhibition of the mitochondrial pyrimidine biosynthesis enzyme dihydroorotate dehydrogenase by doxorubicin and brequinar sensitizes cancer cells to TRAIL-induced apoptosis. Oncogene 33, 3538–3549 [DOI] [PubMed] [Google Scholar]
- 20.Liang Y, Mafuvadze B, Aebi JD, and Hyder SM (2016) Cholesterol biosynthesis inhibitor RO 48–8071 suppresses growth of hormone-dependent and castration-resistant prostate cancer cells. Onco Targets Ther 9, 3223–3232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Mele L, Paino F, Papaccio F, Regad T, Boocock D, Stiuso P, Lombardi A, Liccardo D, Aquino G, Barbieri A, Arra C, Coveney C, La Noce M, Papaccio G, Caraglia M, Tirino V, and Desiderio V (2018) A new inhibitor of glucose-6-phosphate dehydrogenase blocks pentose phosphate pathway and suppresses malignant proliferation and metastasis in vivo. Cell Death Dis 9, 572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Spurr IB, Birts CN, Cuda F, Benkovic SJ, Blaydes JP, and Tavassoli A (2012) Targeting tumour proliferation with a small-molecule inhibitor of AICAR transformylase homodimerization. Chembiochem 13, 1628–1634 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Tagde A, Singh H, Kang MH, and Reynolds CP (2014) The glutathione synthesis inhibitor buthionine sulfoximine synergistically enhanced melphalan activity against preclinical models of multiple myeloma. Blood Cancer J 4, e229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wang K, Luo J, Yeh S, You B, Meng J, Chang P, Niu Y, Li G, Lu C, Zhu Y, Antonarakis ES, Luo J, Huang CP, Xu W, and Chang C (2020) The MAO inhibitors phenelzine and clorgyline revert enzalutamide resistance in castration resistant prostate cancer. Nat Commun 11, 2689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zheng Y, Lin TY, Lee G, Paddock MN, Momb J, Cheng Z, Li Q, Fei DL, Stein BD, Ramsamooj S, Zhang G, Blenis J, and Cantley LC (2018) Mitochondrial One-Carbon Pathway Supports Cytosolic Folate Integrity in Cancer Cells. Cell 175, 1546–1560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Zhuo S, Fan S, and Kaufman S (1996) Effects of depletion of intracellular tetrahydrobiopterin in murine erythroleukemia cells. Exp Cell Res 222, 163–170 [DOI] [PubMed] [Google Scholar]
- 27.Bi X, Han X, Zhang F, He M, Zhang Y, Zhi XY, and Zhao H (2012) Triparanol suppresses human tumor growth in vitro and in vivo. Biochem Biophys Res Commun 425, 613–618 [DOI] [PubMed] [Google Scholar]
- 28.Bronder JL, and Moran RG (2002) Antifolates targeting purine synthesis allow entry of tumor cells into S phase regardless of p53 function. Cancer Res 62, 5236–5241 [PubMed] [Google Scholar]
- 29.Cheng X, Geng F, Pan M, Wu X, Zhong Y, Wang C, Tian Z, Cheng C, Zhang R, Puduvalli V, Horbinski C, Mo X, Han X, Chakravarti A, and Guo D (2020) Targeting DGAT1 Ameliorates Glioblastoma by Increasing Fat Catabolism and Oxidative Stress. Cell Metab 32, 229–242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Cha JH, Yang WH, Xia W, Wei Y, Chan LC, Lim SO, Li CW, Kim T, Chang SS, Lee HH, Hsu JL, Wang HL, Kuo CW, Chang WC, Hadad S, Purdie CA, McCoy AM, Cai S, Tu Y, Litton JK, Mittendorf EA, Moulder SL, Symmans WF, Thompson AM, Piwnica-Worms H, Chen CH, Khoo KH, and Hung MC (2018) Metformin Promotes Antitumor Immunity via Endoplasmic-Reticulum-Associated Degradation of PD-L1. Mol Cell 71, 606–620 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, Li Z, Traugh N, Bu X, Li B, Liu J, Freeman GJ, Brown MA, Wucherpfennig KW, and Liu XS (2018) Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med 24, 1550–1558 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
All data are contained within the article.
