Abstract

Acute graft-versus-host disease (GVHD) is a donor T cell driven complication and the leading cause of non-relapse mortality in patients receiving an allogeneic hematopoietic cell transplantation (allo-HCT). Allogeneic donor T cells eradicate residual leukemia and prevent relapse via the graft-versus-leukemia (GVL) effect and are critical for responding against opportunistic infections post-transplant. Current regimens successful in preventing GVHD are broadly immunosuppressive and come at the cost of increased risk of relapse and/or infection. Therefore, there is an urgent need for new approaches that limit GVHD while retaining GVL responses.

During GVHD, alloreactive T cells boost their energy production through oxidative phosphorylation (OXPHOS) and glycolysis, supporting heightened proliferation and pathogenicity against healthy host tissues. The enzyme dihydroorate dehydrogenase (DHODH), is essential for de novo pyrimidine biosynthesis and for maintaining mitochondrial membrane potential during OXPHOS. Having shown upregulation of DHODH messenger RNA and protein expression in activated human T cells, we evaluated DHODH inhibition, via a small molecule inhibitor HOSU-53, as a therapeutic approach for GVHD. Inhibiting DHODH significantly reduced oxidative metabolism in T cells both during and after activation, while selectively suppressing inflammatory cytokine production in de novo activated, but not previously activated, T cells. In a xenogeneic model, HOSU-53 treatment limited GVHD severity, decreased pathogenic Th1 and Th17 response, and preserved beneficial GVL effects. Altogether, we identify DHODH inhibition as an innovative treatment strategy in allo-HCT recipients to reduce GVHD severity and retain effective GVL response.

Introduction

Over 8,000 allogeneic hematopoietic cell transplants (allo-HCT) are performed annually in the U.S. and represent the only curative therapy for many high-risk hematological malignancies.1–5 In patients with acute myeloid leukemia (AML), myelodysplastic syndromes, and acute lymphoblastic leukemia, an allo-HCT replaces the dysfunctional, malignant hematopoietic system with stem cells from an HLA-matched healthy donor while donor immune cells eliminate residual disease via the beneficial graft-versus-leukemia (GVL) effect.6–8 Allogeneic T cells can also recognize the recipient tissues as foreign through major and/or minor HLA mismatching, causing a severe immunological complication known as acute graft-versus-host disease (GVHD).6,7,9–12 Despite advances in donor matching, conditioning regimens, and prophylactic strategies, between 30% and 70% of allo-HCT patients develop acute GVHD which remains a leading cause of non-relapse mortality.13,14 The recipients’ conditioning regimen causes cellular apoptosis and breach of the intestinal barrier releasing pathogen- and damage- associated molecular patterns as well inflammatory chemokines and cytokines.9,15 In this inflammatory milieu, recognition of recipient allo-antigens by donor T cells results in activation, expansion, and migration of alloreactive donor T cells to tissues such as the skin, liver, and GI tract, with severe cases resulting in end-organ damage and possible death of the recipient.9,15

Prophylactic regimens for GVHD commonly consist of combinations of calcineurin inhibitors and antimetabolites.16,17 Recent clinical trials have shown that using post-transplant cyclophosphamide (PTCy) as prophylaxis reduces the occurrence of both acute and chronic GVHD, without a significant impact on the relapse rate.16,18–20 Extensive preclinical21–23 and clinical24 investigations into the role of CD28 co-stimulation in T cell alloreactivity have resulted in FDA approval for abatacept, a selective T cell costimulatory modulator. Abatacept has been effective in reducing the incidence of severe Grade III to IV acute GVHD, although it does not substantially improve the rates of chronic GVHD or the occurrence of relapse and viral infections.25–28 Despite continuous improvements in GVHD prophylaxis, approximately 52% of allogeneic hematopoietic stem cell transplant (allo-HSCT) recipients still develop Grade II to IV GVHD, subsequent treatments remain suboptimal, and 15% of patients experience severe GVHD with poor outcomes.9,29–34

During GVHD, alloreactive T cells upregulate both oxidative phosphorylation (OXPHOS) and glycolysis leading to increased proliferation and cytolytic activity against healthy host tissues.35–39 Targeting these metabolic processes post allogeneic transplant has shown promise in alleviating acute GVHD while preserving beneficial GVL effects in mouse models.36,40,41 In vivo, T cells deficient in mitochondrial electron transport chain (ETC) complexes III or IV fail to proliferate after antigen exposure.42–45 Interestingly, impairing ETC function does not interfere with thymic development42,43 as these T cells meet their energy demands through glycolysis and metabolite generation through other redundant mechanisms. Therefore, targeting mitochondrial membrane potential and ETC function could be a viable strategy to mitigate GVHD without impairing GVL function of the donor T cells.

The rate-limiting enzyme dihydroorate dehydrogenase (DHODH) is involved in de novo pyrimidine biosynthesis and in maintaining mitochondrial membrane potential for the ETC, processes vital for T cell proliferation and effector function. DHODH donates an electron to ubiquinone causing its’ transfer to ubiquinol. ETC complex III oxidizes ubiquinol back to ubiquinone, maintaining function of complexes I and II.46 Inhibition of DHODH has been identified as a promising therapeutic strategy for T cell mediated autoimmune disorders such as multiple sclerosis (MS)47,48 and amyotrophic lateral sclerosis49 as well as for treating hematological malignancies such as Acute Myeloid Leukemia.50 In MS, DHODH promotes expansion of T cells bearing high-affinity TCRs, and inhibiting DHODH function, vis-a-vis teriflunomide, corrects metabolic disturbances in T cells, subsequently alleviating disease.51 HOSU-53 is a potent DHODH inhibitor, with nanomolar inhibitory capacity and pharmaceutical properties enabling once daily oral dosing. Most importantly, HOSU-53 has demonstrated therapeutic efficiency in murine models of AML.52 However, the relevance of DHODH function and therapeutic potential within the context of GVHD is largely unknown. In this manuscript, we evaluated DHODH inhibition, using the pharmacological inhibitor (HOSU-53), as a novel strategy to mitigate both acute GVHD and relapse in xenogeneic models of GVHD and leukemia.

Materials and methods

Mice

NSG mice were purchased from the OSUCCC Target Validation Shared Resource (Columbus, Ohio, USA). For transplant experiments, recipient mice were between 12 and 16 wks of age. All animal studies were conducted in accordance with the rules and regulations of the Institutional Animal Care and Use Committee at OSU.

Agilent seahorse ATP rate assay

Metabolic assays were performed on the Agilent Seahorse XFe96 following manufacturer’s instructions and using all recommended reagents supplied by the manufacturer. Briefly, plates were coated with Cell-Tak at 22.4 µg/ml (Corning 354240) and neutralized with 0.1M sodium bicarbonate, incubated for 20 minutes at room temperature, then washed twice with sterile water. T cells were suspended at 40x105 cells/mL in XF Assay Media [Seahorse XF DMEM Media, pH 7.4 (Agilent 103575-100), 10 mM Seahorse XF Glucose (1.0 M stock, Agilent 103577-100), 1 mM Seahorse XF Pyruvate (100 mM stock, Agilent 103578-100), 2 mM Seahorse XF L-Glutamine (100 mM stock, Agilent 103579-100)] and plated at 200,000 cells/well in the coating 96-well plate. Cells were adhered to the plate by centrifugation prior to the addition of more XF Assay Media to bring wells to a total volume of 180 µl. For the Mito Stress Test (Agilent 103015-100), Oligomycin, FCCP, and Rotenone + Antimycin A were suspended in XF Assay Media to a concentration of 15 µM, 5 µM, and 5 µM, respectively, and loaded into the hydrated sensor cartridge at volumes of 20 µl, 22 µl, and 25 µl, respectively. For the Glyco Stress Test (Agilent 103020-100), Glucose, Oligomycin, and 2-DG were suspended in XF Assay Media to a concentration of 10 µM, 1 µM, and 50 µM, respectively, and loaded into the hydrated sensor cartridge at volumes of 20 µL , 22 µL, and 25 µL, respectively. Assays was performed using the Wave Software (Agilent) and data analyzed using Seahorse Analytics (Agilent).

qRT-PCR

RNA was isolated from 0.5-2x106 cells using Trizol (Invitrogen 15596026) and Phasemaker Tubes (Invitrogen A33248) and cDNA was made from 500 ng of RNA using the RevertAid RT Reverse Transcription Kit (Thermo Scientific K1691), following manufacturer’s instructions. qRT-PCR was performed using TaqMan Real-Time PCR Assays (Applied Biosystems) and TaqMan Universal PCR Master Mix (Applied Biosystems 4304437) with 1 µl cDNA (5 µl of a 1:5 dilution loaded per well). Individual assays used are: DHODH (Hs00361406_m1), Hprt1 (Hs02800695_m1), SDHA (Hs00188166_m1), and UBE2D2 (Hs00366152_m1).

Western blot

Cells were lysed in RIPA buffer (Pierce) with protease inhibitor cocktail (Millipore: 539131-1VL) Protein was quantified using the Pierce Rapid Gold BCA Protein Assay Kit (Thermo Scientific A53225). 20 µg of total protein was loaded per well and transferred to nitrocellulose membrane. Primary antibodies for DHODH (CST no. 26381) and Vinculin (CST no. 13901) were incubated at 4 °C overnight and secondary antibodies were incubated for 1 hour at room temperature (IRDye 680RD goat anti-rabbit (Li-Cor 926-68071) and IRDye 800CW donkey anti-mouse (Li-Cor 926-32212), both 1:20,000). Fluorescence was detected using the Li-Cor Odyssey CLx and band intensity quantification was performed using the ImageStudio software (Li-Cor).

Cell culture and flow cytometry

Human T cells were isolated from buffy coats (Versiti Indiana) using the Pan T cell Isolation Kit (Miltenyi Biotec). Cells were stimulated with CD3/CD28 Dynabeads (Invitrogen) for 48 to 72 h ± HOSU-53, followed by incubation for 5 hours with Cell Stimulation Cocktail (eBioscience) and Protein Transport Inhibitor (Thermo Fisher Scientific). Cells were then stained with LIVE/DEAD fixable cell viability dye (Thermo Fisher Scientific) and surface antibodies, permeabilized and fixed, followed by staining with intracellular antibodies and analyzed within 24 h. For post activation studies, human T cells were stimulated first with CD3/CD28 Dynabeads for 72 h. Beads were subsequently removed, and cells rested and expanded with rhIL-2 (Peprotech) ± HOSU-53 for 72 h. On the day before metabolic/flow cytometry analysis, cells were restimulated with CD3/CD28 Dynabeads overnight. Data was acquired using a spectral Cytek Aurora flow cytometer, and analysis performed using FlowJo (Tree Star).

Xenogeneic GVHD model

Mice were transplanted under standard protocols approved by the University Committee on Use and Care of Laboratory Animals at OSU. Only age- and sex-matched mice were used for transplant experiments. Briefly, NSG mice were irradiated with 100 cGy one day before transplant. Fresh human PBMCs (1.7-2.0 x107) were administered on the day of transplant by tail-vein injection. Recipients were treated with vehicle or DHODH inhibitor HOSU-53 (10 mg/kg), administered by oral gavage twice weekly starting at day +1 post-transplant until the end of the study.

Clinical assessment of acute GVHD

Recipient mice were weighed 2 to 4 times per week and monitored daily for clinical signs of acute GVHD and survival. A scoring method adapted and modified from Cooke et al.53 was used to assess clinical changes associated with acute GVHD. Briefly, this scoring system incorporates 5 clinical parameters: weight loss, posture (hunching), activity, fur texture, and skin integrity. Individual mice were ear tagged and graded (on a scale from 0 to 10) 3 times a week. Mice who reached an acute GVHD score of more than or equal to 7 were euthanized and their tissues harvested. A separate cohort of mice were euthanized on day 40 post-transplant and single cell suspensions from the spleen and liver were generated via manual homogenization and liver dissociation kit, (Miltenyi no. 130105807) respectively, and assessed by flow cytometry. The histological intensity of acute GVHD in different liver sections was assessed on a 28-point scale by evaluating 7 parameters comprising portal tract inflammation, bile duct injury, vascular endotheliitis, periportal, or limiting plate necrosis, lobular necro-inflammatory activity, zonal necrosis, and sinusoidal lymphocytosis.54

Xenogeneic GVL model

NSG recipients were irradiated (100 cGy) on day −1. Firefly luciferase and GFP transduced MOLM-13 acute myeloid leukemia cells (0.5 × 106) were injected intravenously on day 0. Human PBMCs (1.7–2.0 ×107) were injected intravenously on day +1. Treatment groups included vehicle and HOSU-53 10 mg/kg, administered by oral gavage twice weekly starting day +2 post-transplant. Molm-13 and human PBMCs alone served as the control groups. Molm-13-induced death was defined by hind-limb paralysis. GVHD death was defined as the absence of leukemia and presence of clinical and histopathological signs of GVHD.

In vivo imaging

Xenogen IVIS imaging system (Caliper Life Sciences) was used for live animal imaging. Mice were anesthetized using 1.5% isofuorane (Piramal Healthcare). XenoLight RediJect D-Luciferin Ultra Bioluminescent Subtrate (150 mg/kg body weight; 30 mg/ml in PBS; Perkin Elmer) was injected intraperitoneally and IVIS imaging was performed 7 to 10 min after substrate injection. Whole body bioluminescent signal intensity was determined using IVIS Living Image software v4.3.1 (Caliper Life Sciences), and pseudo-color images overlaid on conventional photographs are shown. Data were analyzed and presented as photon counts per area.

Degranulation assay

CD8 T cell degranulation was measured by intracellular production of IFN-γ and CD107a in response to ex vivo PMA/ionomycin stimulation. Splenocytes harvested from vehicle and HOSU-53 treated mice were stimulated and stained for intracellular cytokines as described above. Donor T cells were stained with human CD45 for identification. Cells were analyzed by flow cytometry.

Statistical analysis

Survival data were analyzed using Kaplan-Meier curves and statistics analyzed via log-rank test. Student unpaired t-test was used for all statistical analysis except where indicated. One-way ANOVA with Dunnet post-hoc test was used for comparisons with greater than 2 groups. For estimating statistical significance in clinical scores using multiple t-tests over time, P-values were adjusted using the 2-stage linear step-up procedure of Benjamini, Krieger, and Yekutieli.55 Data represent mean ± SD. All analyses were performed using GraphPad Prism 9.0. *P < 0.05, **P < 0.01, ***P < 0.001, NS non-significant.

Results

DHODH inhibition reduces T cell proliferation and function during de novo activation

Activated human T cells upregulate DHODH as evidenced by a significant increase in both DHODH mRNA (Fig. 1A) and protein expression (Fig. 1B, C) following CD3/CD28 stimulation. To investigate the impact of DHODH inhibition on T cell proliferation and viability, cell-trace violet labeled T cells were stimulated with CD3/CD28 beads in the presence of a range of HOSU-53 (0–1,000 nM). We observed a 50% reduction in T cell proliferation at 10 nM and 100 nM (Fig. 1D, E); however, we also observed reduction in T cell viability at 100 nM (Fig. 1F). Thereby, we selected 10 nM as the optimal concentration for further in vitro studies.

DHODH Inhibition reduces human T cell proliferation in vitro. T cells were isolated from healthy human donors and stimulated with CD3/CD28 Dynabeads for 48 h. (A) Fold Change in DHODH mRNA expression by qRT-PCR between unstimulated (US) and stimulated human T cells (n = 3); (B) representative western blot of DHODH protein expression in one donor, with quantification from multiple donors shown in (C) (n = 4). (D) Cell trace violet–labeled (CTV-labeled) human T cells were stimulated with CD3/CD28 Dynabeads for 72 h in the presence of increasing concentrations of DHODH inhibitor HOSU-53. CTV dilution of one representative donor is shown. (E) Percent CD3+ T cell proliferation, where each symbol represents an individual donor (n = 6). (F) T cells isolated from healthy human donors were activated with CD3/CD28 Dynabeads, with and without increasing concentrations of HOSU-53, for 72 h. Followed by flow cytometric analysis of Annexin/PI expression (n = 4). *P < 0.05 **P < 0.01 ***P < 0.001 ****P < 0.0001. Student paired t-test determined statistical significance.
Figure 1.

DHODH Inhibition reduces human T cell proliferation in vitro. T cells were isolated from healthy human donors and stimulated with CD3/CD28 Dynabeads for 48 h. (A) Fold Change in DHODH mRNA expression by qRT-PCR between unstimulated (US) and stimulated human T cells (n = 3); (B) representative western blot of DHODH protein expression in one donor, with quantification from multiple donors shown in (C) (n = 4). (D) Cell trace violet–labeled (CTV-labeled) human T cells were stimulated with CD3/CD28 Dynabeads for 72 h in the presence of increasing concentrations of DHODH inhibitor HOSU-53. CTV dilution of one representative donor is shown. (E) Percent CD3+ T cell proliferation, where each symbol represents an individual donor (n = 6). (F) T cells isolated from healthy human donors were activated with CD3/CD28 Dynabeads, with and without increasing concentrations of HOSU-53, for 72 h. Followed by flow cytometric analysis of Annexin/PI expression (n = 4). *P < 0.05 **P < 0.01 ***P < 0.001 ****P < 0.0001. Student paired t-test determined statistical significance.

In corroboration with the proliferation studies, we observed a decrease in Ki67 expression across both CD4+ and CD8+ compartments of T cells following HOSU-53 treatment (Fig. 2A). Furthermore, DHODH inhibition also significantly reduced IFN-γ and TNF-α production, indicating decreased inflammatory function (Fig. 2B, C). Given DHODH localization in the mitochondria, and its’ contribution to mitochondrial membrane potential,50 we assessed OXPHOS and glycolysis in T cells treated with HOSU-53. Inhibition of DHODH decreased mitochondrial respiration, diminishing both maximal respiration and spare respiratory capacity (Fig. 2D, E), while also significantly reducing glycolysis and glycolytic capacity (Fig. 2F, G). To test whether the observed metabolic impairment was independent of the role of DHODH in de novo pyrimidine synthesis, we conducted a uridine rescue assay. Our results show that HOSU-53 mediated reductions in OXPHOS and glycolysis were rescued in the presence of uridine supplementation (Fig. S1).

DHODH inhibition reduces de novo activated T cell inflammatory cytokine production and oxidative metabolism in vitro. T cells isolated from healthy human donors were activated with CD3/CD28 Dynabeads with and without 10 nM HOSU-53 for 72 h, followed by flow cytometric analysis of (A) Ki67, (B) IFN-γ, and (C) TNF-α expression (n = 6). (D) Oxygen consumption rate (OCR) of T cells from one representative donor during mitostress test with bar graphs (E) of maximal respiration and spare respiratory capacity. (F) Extracellular acidification rate (ECAR) of T cells during glycolytic stress test with bar graphs of (G) glycolysis and glycolytic capacity of T cells (n = 4 to 5 independent donors). *P < 0.05 **P < 0.01 ***P < 0.001 ****P < 0.0001.
Figure 2.

DHODH inhibition reduces de novo activated T cell inflammatory cytokine production and oxidative metabolism in vitro. T cells isolated from healthy human donors were activated with CD3/CD28 Dynabeads with and without 10 nM HOSU-53 for 72 h, followed by flow cytometric analysis of (A) Ki67, (B) IFN-γ, and (C) TNF-α expression (n = 6). (D) Oxygen consumption rate (OCR) of T cells from one representative donor during mitostress test with bar graphs (E) of maximal respiration and spare respiratory capacity. (F) Extracellular acidification rate (ECAR) of T cells during glycolytic stress test with bar graphs of (G) glycolysis and glycolytic capacity of T cells (n = 4 to 5 independent donors). *P < 0.05 **P < 0.01 ***P < 0.001 ****P < 0.0001.

Our studies so far investigated the impact of DHODH inhibition when used concurrently with T cell activation. To investigate whether DHODH inhibition could modulate the function of previously activated T cells, we first stimulated T cells and then treated with HOSU-53. Interestingly, in this setting we found that DHODH inhibition reduced only OXPHOS (Fig. 3A, B), with no impact on glycolysis and glycolytic capacity (Fig. 3C, D). Further, there was no difference in Ki67 expression, IFN-γ, or TNF-α cytokine production in previously activated T cells (Fig. 3E–G). This suggests that DHODH inhibition preferentially targets T cell function during de novo activation without impacting the phenotype of already activated T cells.

DHODH inhibition blocks oxidative metabolism of previously activated T cells without affecting inflammatory cytokine production. T cells isolated from healthy human donors were activated with CD3/CD28 Dynabeads for 72 h. Beads are subsequently removed and cells expanded with 10 ng/ml rhIL-2 with and without 10 nM HOSU-53 for an additional 72 h. On day prior to analysis, cells were restimulated with CD3/CD28 Dynabeads overnight. Metabolic stress tests measuring (A) OCR of T cells from one representative donor during mitostress test with bar graphs of (B) maximum respiration and spare respiratory capacity. (C) Extracellular acidification rate of T cells during glycolytic stress test measuring (D) glycolysis and glycolytic capacity of T cells (n = 6 independent donors). Flow cytometric analysis of (E) Ki67, (F) IFN-γ, and (G) TNF-α expression (n = 6). *P < 0.05 **P < 0.01 ***P < 0.001 ****P < 0.0001.
Figure 3.

DHODH inhibition blocks oxidative metabolism of previously activated T cells without affecting inflammatory cytokine production. T cells isolated from healthy human donors were activated with CD3/CD28 Dynabeads for 72 h. Beads are subsequently removed and cells expanded with 10 ng/ml rhIL-2 with and without 10 nM HOSU-53 for an additional 72 h. On day prior to analysis, cells were restimulated with CD3/CD28 Dynabeads overnight. Metabolic stress tests measuring (A) OCR of T cells from one representative donor during mitostress test with bar graphs of (B) maximum respiration and spare respiratory capacity. (C) Extracellular acidification rate of T cells during glycolytic stress test measuring (D) glycolysis and glycolytic capacity of T cells (n = 6 independent donors). Flow cytometric analysis of (E) Ki67, (F) IFN-γ, and (G) TNF-α expression (n = 6). *P < 0.05 **P < 0.01 ***P < 0.001 ****P < 0.0001.

DHODH inhibition reduces in vivo GVHD severity

To evaluate the therapeutic potential of DHODH inhibition, we utilized a xenogenic model of acute GVHD wherein NSG mice were transplanted with human PBMCs and treated with HOSU-53 or vehicle control. DHODH inhibition via HOSU-53 significantly extended survival (Fig. 4A, median survival 56 vs. 40 days, HOSU-53 vs. control, P < 0.01) and decreased GVHD severity, as observed by reduced clinical scores (Fig. 4B). We found that although mice under DHODH inhibition eventually succumbed to GVHD, HOSU-53 treatment significantly reduced liver damage compared to vehicle treatment alone (Fig. 4C, D). Furthermore, HOSU-53 also reduced pro-inflammatory IFN-γ and TNF-α cytokine production by donor CD4+ T cells taken from the spleen (Fig. S2A, B). To further investigate this pathology, we conducted a separate experiment using the same xenogeneic GVHD model, but created single cell suspensions of whole liver tissue at day 40 post allo-HCT for FACS analysis. In this case, we found decreased absolute numbers of inflammatory Th1, Th17, and TNF-α+ CD4 T cells infiltrating the liver (Fig. 4E–G) as well as a decrease in Treg numbers (Fig. 4H). To investigate whether DHODH inhibition impacted antigen presentation, we evaluated MHCII expression on donor and recipient splenic antigen presenting cells and found no differences between treatment cohorts (Fig. S3AC). Altogether, these data indicate that DHODH inhibition in human T cells provides protection from acute GVHD disease progression and lethality in vivo.

DHODH inhibition improves recipient survival during xenogeneic GVHD. NSG mice were transplanted as described in Methods and treated with vehicle or HOSU-53 at 10 mg/kg starting at day 1 and twice weekly thereafter. (A) Survival curve (n = 8 to 9 per group) with data combined from 2 independent donor transplants. (B) Average GVHD clinical scores of mice treated with vehicle or HOSU-53 at 10 mg/kg (n = 4 per group). One representative transplant is shown. (C) Livers were harvested at time of death, formalin fixed, paraffin embedded and liver sections stained with hematoxylin and eosin. (D) Histopathological scores of liver sections determined as described in Methods. (E–H), Liver tissues were harvested at day 40 and analyzed via FACS; data are combined from 2 independent donor transplants. Bar graphs show enumeration of donor (E) Th1 (IFN-γ+, TBET+), (F) Th17 (IL-17+, Ror-γt+), (G) TNF-α+ CD4, and (H) FoxP3+, CD25+ CD4+ Treg cells in the liver. *P < 0.05, **P < 0.01. *** P < 0.001, *** P < 0.0001.
Figure 4.

DHODH inhibition improves recipient survival during xenogeneic GVHD. NSG mice were transplanted as described in Methods and treated with vehicle or HOSU-53 at 10 mg/kg starting at day 1 and twice weekly thereafter. (A) Survival curve (n = 8 to 9 per group) with data combined from 2 independent donor transplants. (B) Average GVHD clinical scores of mice treated with vehicle or HOSU-53 at 10 mg/kg (n = 4 per group). One representative transplant is shown. (C) Livers were harvested at time of death, formalin fixed, paraffin embedded and liver sections stained with hematoxylin and eosin. (D) Histopathological scores of liver sections determined as described in Methods. (E–H), Liver tissues were harvested at day 40 and analyzed via FACS; data are combined from 2 independent donor transplants. Bar graphs show enumeration of donor (E) Th1 (IFN-γ+, TBET+), (F) Th17 (IL-17+, Ror-γt+), (G) TNF-α+ CD4, and (H) FoxP3+, CD25+ CD4+ Treg cells in the liver. *P < 0.05, **P < 0.01. *** P < 0.001, *** P < 0.0001.

HOSU-53 does not impair the beneficial GVL effect in allo-HCT

Relapse prevention serves as the main purpose of allogenic transplants in leukemia-bearing patients. We therefore investigated whether HOSU-53 preserved anti-leukemic effect of donor T cells against AML. In vitro, HOSU-53 treatment retained anti-tumor T cell efficacy (Fig. S4B). To investigate this effect in vivo, we used a xenogeneic model of GVL wherein NSG mice were infused with a GFP+ luciferase transduced FLT3-ITD+ MOLM-13 AML cell line along with PBMCs and subsequently treated with HOSU-53 or vehicle. We observed an overall improvement in median survival in mice receiving both PBMCs and HOSU-53 treatment (Fig. 5A, B, median survival 29 vs. 24 d, HOSU-53 vs. control, P < 0.0001). To help assess for cause of death, tumor burden was measured via flow cytometric evaluation of GFP+ MOLM-13 cells in the spleen at end of study. This analysis showed comparable levels of leukemic cells in the spleen in mice that received either MOLM-13 alone or MOLM-13 plus HOSU-53 (∼70% GFP+ MOLM-13 cells). In contrast, mice receiving PBMCs, with or without HOSU-53 treatment, eradicated GFP+ MOLM-13 tumor cells equally, showing retention of beneficial GVL effects in the presence of HOSU-53 (Fig. 5B, C). DHODH inhibition also retained IFN-γ and TNF-α production by CD8+ and CD4+ T cells, likely contributing to their anti-tumor function (Fig. 5D–G).

DHODH inhibition retains beneficial GVL effects. Xenogeneic GVL transplant was performed as described in Methods, with NSG recipient mice treated with vehicle or HOSU-53 at 10 mg/kg starting at day 2, then twice weekly (n = 7 to 15 per group). (A) Survival curve. (B) Whole-body bioluminescent signal intensity of recipient mice (n = 3 per cohort) on indicated days. (C–G), At time of death, spleens were harvested and analyzed via FACS for C) GFP+ MOLM-13+ cell population to evaluate leukemic burden, as well as IFN-γ production by human donor D) CD4+, E) CD8+ and TNF-α production in F) CD4+ and G) CD8+ T cells. *P < 0.05, **P < 0.01. *** P < 0.001, *** P < 0.0001.
Figure 5.

DHODH inhibition retains beneficial GVL effects. Xenogeneic GVL transplant was performed as described in Methods, with NSG recipient mice treated with vehicle or HOSU-53 at 10 mg/kg starting at day 2, then twice weekly (n = 7 to 15 per group). (A) Survival curve. (B) Whole-body bioluminescent signal intensity of recipient mice (n = 3 per cohort) on indicated days. (C–G), At time of death, spleens were harvested and analyzed via FACS for C) GFP+ MOLM-13+ cell population to evaluate leukemic burden, as well as IFN-γ production by human donor D) CD4+, E) CD8+ and TNF-α production in F) CD4+ and G) CD8+ T cells. *P < 0.05, **P < 0.01. *** P < 0.001, *** P < 0.0001.

Discussion

Agents that prevent GVHD without increasing the risk of relapse remain elusive in the post-allo-HCT landscape. Using HOSU-53, a novel small-molecule inhibitor with favorable oral bioavailability, we show that DHODH inhibition prevents GVHD while retaining the GVL effects in human T cells. Our results help to better understand the therapeutic effects of pyrimidine synthesis inhibitors post allo-HCT and we anticipate that the knowledge gained in this preclinical work will provide further support for the clinical translation of DHODH inhibitors that selectively influence effector T cell function.

Nucleotide synthesis inhibitors such as mycophenolate mofetil and methotrexate are commonly used as GVHD prophylaxis strategies.56 Despite this treatment, infections and GVHD are two of the most common reasons for hospitalization of allo-HCT recipients,55 possibly due to the broad immunosuppressive action of these agents with nonspecific reductions in both effector and memory T cells.57 We initially became interested in studying the DHODH pathway due to selective retention of infectious immune response in MS patients who received teriflunomide, a DHODH inhibitor.58 Inhibiting DHODH in MS mouse models showed decreased clonal expansion and effector function of high-affinity T cells.47,48 Thus, we posited that inhibition of DHODH, and its’ differential impact on T cell subsets,59–61 might be an effective strategy to mitigate GVHD while retaining GVL responses.

Our results demonstrate that DHODH expression is upregulated in activated T cells and that disruption of DHODH modulates multiple components of T cell effector function, including proliferation and inflammatory TNF-α and IFN-γ cytokine production during de novo activation. Furthermore, DHODH inhibition results in diminished T cell metabolic function, with decreased mitochondrial respiration and glycolysis. Interestingly, T cells that receive DHODH inhibition post-activation still retain proliferative capability, as evidenced by Ki67 expression, as well as inflammatory TNF-α and IFN-γ production, despite a major lesion in OXPHOS. These results suggest that DHODH is selectively integral to the development of robust effector T cells, consistent with results in the literature where DHODH inhibition selectively blocked T differentiation into an effector phenotype.61 Interestingly, we observed a rescue of OXPHOS and glycolytic function in T cells treated with HOSU-53 and supplemented with uridine, suggesting that the impact of DHODH inhibition on oxidative metabolism is downstream of nucleotide starvation, in corroboration with previously published research.51

While it is known alloreactive T cells rely heavily on OXPHOS and aerobic glycolysis during early activation, fully understanding the metabolic processes within GVHD remains a work in progress. Previous work has shown that early glucose uptake by alloreactive T cells leads to enhanced effector function and tissue damage. These T cells rely on increased mitochondrial respiration and are vulnerable to ETC manipulation.35 Molecular inhibition of glycolysis selectively suppresses alloreactive T cells, and genetic ablation of glucose uptake in donor T cells prevents GVHD while retaining GVL.40 Furthermore, Inhibiting ATPase in mitochondrially active T cells improves survival, lowers clinical scores, and decreases lymphocytic infiltration into target organs in GVHD.41 Notably, under DHODH inhibition, we observed decreased Th1 (CD4+TBET+IFN-γ+), Th17 (CD4+Rorγt+IL17+) and CD4+TNF-α T cells within our GVHD model. Furthermore, Treg (Foxp3+, CD25hi) cells, well known to rely heavily on OXPHOS for their metabolic needs,62–64 were similarly diminished following HOSU-53 treatment, suggesting that increased tolerance was not likely to be the mechanism underlying the improved survival benefit. Interestingly, we observed increased CD8+IFN-γ+ T cells within our GVL model. Congruently, others have commented on the differential metabolic profiles between CD4+ and CD8+ T cells, and metabolic manipulation leading to diminished GVHD pathogenesis without significantly impacting GVL.65 However, these previous in-vivo research has been limited to murine models, without much investigation on impact on human T cells. Here, using a human PBMC into NSG model of xenogeneic GVHD, we identify DHODH as a contributor to effector function in human T cells during acute GVHD pathogenesis. Furthermore, this metabolic manipulation retains the necessary GVL responses post allo-HCT. HOSU-53 treatment alone is insufficient at clearing tumor at the administered dose of 10 mg/kg twice weekly, but it also does not disrupt the ability of PBMCs to clear allogeneic tumor cells. To note, this 10 mg/kg twice weekly is a much lower dose than what has been previously reported as required for maximal survival in an AML model (10 mg/kg daily or 30 mg/kg twice weekly).52 This infers that the survival benefit seen with concurrent PBMC and HOSU-53 administration is likely attributable to a retained GVL effect versus any direct anti-leukemic effect of DHODH inhibition.

Recently, allo-HCT phase II clinical trials evaluating efficacy of naïve T cell depleted allografts revealed very low incidence of acute GVHD along with no chronic GVHD and no apparent risk of relapse or non-relapse mortality.66 Supporting these data, murine models of allo-HCT showed that in the absence of naïve T cells, CD4+CD25- memory T cells do not cause GVHD59 while CD8+ memory and naïve T cells have equivalent GVL capabilities.60 Concurrently, DHODH was found to be integral to the development of robust effector T cells. A caveat of the xenogeneic GVHD model is that it is not optimal for evaluating memory responses, however, previously published research has shown that inhibition of DHODH in patients with MS selectively blocked CD8+ T cell differentiation into an effector phenotype while retaining memory T cell responses against infectious agents.61 Therefore, pharmacologically inhibiting DHODH could selectively inhibit naïve alloreactive T cell activation and induce differentiation into memory T cells thereby limiting GVHD severity while preserving GVL.

In conclusion, HOSU-53 is a potent DHODH inhibitor, impacting T cell metabolism and immune function. DHODH inhibition post-transplant increases recipient survival and decreases GVHD severity, without impairing human T cell anti-leukemic function in a xenogeneic model. Our results, along with the reported direct anti-leukemic effects of HOSU-53,51 provide a strong rationale for the use of DHODH inhibition post allo-HCT to prevent acute GVHD while minimizing risk of relapse.

Acknowledgments

This research was made possible through resources, expertise, and support provided by the Pelotonia Institute for Immuno-Oncology (PIIO), which is funded by the Pelotonia community and the OSUCCC. We thank the PIIO and the Immune Monitoring and Discovery Platform for spectral flow cytometry and Seahorse assays. We thank the Target Validation Shared Resource (TVSR) and the Drug Discovery Shared Resource (DDSR) at the Ohio State University Comprehensive Cancer Center for providing the NSG mice and HOSU-53, respectively, used in the preclinical studies described herein. TVSR and MCSR are NCI supported shared resources through the OSUCCC grant P30 CA016058. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institute of Health.

Author contributions

R.K. performed T cell isolations, seahorse assays, western blots, in vitro and ex vivo FACS analysis, in vivo GVHD models, analyzed the data and wrote the manuscript. K.M.B. and L.N.C. performed the T cell isolations, seahorse assays, ex-vivo FACS analysis, real-time PCR, and in vivo GVHD and GVL models. N.S and Y.G. performed T cell isolations, in-vitro T cell assays and in-vivo imaging. C.B. and S.V. provided the HOSU-53 drug. C.B., C.A.B., O.A.E, T.G, E.H, H.K.C., and J.C.B. provided ideas, discussion, and edited the manuscript. P.R designed the study, supervised research, analyzed and interpreted the data, and edited the manuscript.

Supplementary material

Supplementary material is available at The Journal of Immunology online.

Funding

This work was supported by The OSUCCC Drug Development Institute, the Paula and Rodger Riney Foundation, OSU CCC and Division of Hematology start-up funds, NIH R01CA252469, R01HL163849, American Cancer Society Research Scholar Grant RSG-22-053-01-IBCD (P.R.); NIH T32CA090223 fellowship to K.B. Research reported in this publication was supported by the Ohio State University Comprehensive Cancer Center and the National Institutes of Health under grant number P30 CA016058.

Conflicts of interest

J.C.B., T.E.G., O.A.E., E.K.H., C.B., and S.V. are inventors on the patent for HOSU-53. H.K.C. has served on advisory boards or consulting for Incyte, Sanofi, and Actinium and research support from Opna. The remaining authors declare no competing financial interests.

Data availability

The data underlying this article are available in the article and in its online supplementary material.

References

1

Hamilton
BK.
 
Updates in chronic graft-versus-host disease
.
Hematology (United States)
.
2021
;
2021
:
648
654
.

2

Auletta
J
,
Kou
J
,
Chen
M
,
Shaw
B.
Current use and outcome of hematopoietic stem cell transplantation: CIBMTR US summary slides 2021.
2021
. https://cibmtr.org/Files/Summary-Slides—Reports/The-US-Summary-Slides-2021-v2—web-version.pptx

3

Choi
SW
,
Reddy
P.
 
Current and emerging strategies for the prevention of graft-versus-host disease
.
Nat Rev Clin Oncol
.
2014
;
11
:
536
547
.

4

Holtan
SG
,
Marcelo
P
,
Weisdorf
DJ.
 
Acute graft-versus-host disease: a bench-to-bedside update
.
Blood
.
2014
;
124
:
363
373
.

5

Poiré
X
 et al.  
Hematopoietic stem cell transplantation for adult patients with isolated NPM1 mutated acute myeloid leukemia in first remission
.
Am J Hematol
.
2019
;
94
:
231
239
.

6

Perkey
E
,
Maillard
I.
 
New insights into graft-versus-host disease and graft rejection
.
Annu Rev Pathol
 
2018
;
13
:
219
245
.

7

Socié
G
,
Kean
LS
,
Zeiser
R
,
Blazar
BR.
 
Insights from integrating clinical and preclinical studies advance understanding of graft-versus-host disease
.
J Clin Invest
.
2021
;
131
.

8

Dickinson
AM
 et al.  
Graft-versus-leukemia effect following hematopoietic stem cell transplantation for leukemia
.
Front Immunol
.
2017
;
8
:
496
.

9

Welniak
LA
,
Blazar
BR
,
Murphy
WJ.
 
Immunobiology of allogeneic hematopoietic stem cell transplantation
.
Annu Rev Immunol
.
2007
;
25
:
139
170
.

10

Nassereddine
S
,
Rafei
H
,
Elbahesh
E
,
Tabbara
I.
 
Acute graft versus host disease: a comprehensive review
.
Anticancer Res
.
2017
;
37
:
1547
1555
.

11

Zeiser
R.
 
Introduction to a review series on pathophysiology and treatment of acute GVHD
.
Blood
.
2020
;
136
:
375
376
.

12

Malard
F
,
Holler
E
,
Sandmaier
BM
,
Huang
H
,
Mohty
M.
 
Acute graft-versus-host disease
.
Nat Rev Dis Primers
.
2023
;
9
:
27
.

13

Ferrara
JLM
,
Levine
JE
,
Reddy
P
,
Holler
E.
 
Graft-versus-host disease
.
Lancet
.
2009
;
373
:
1550
1561
.

14

Kumar
R
 et al.  
Comparing outcomes with bone marrow or peripheral blood stem cells as graft source for matched sibling transplants in severe aplastic anemia across different economic regions
.
Biol Blood Marrow Transplant
.
2016
;
22
:
932
940
.

15

Singh
AK
,
McGuirk
JP.
 
Allogeneic stem cell transplantation: a historical and scientific overview
.
Cancer Res
.
2016
;
76
:
6445
6451
.

16

Jamy
O
,
Zeiser
R
,
Chen
YB.
 
Novel developments in the prophylaxis and treatment of acute GVHD
.
Blood
.
2023
;
142
:
1037
1046
.

17

Gooptu
M
,
Antin
JH.
 
GVHD Prophylaxis 2020
.
Front Immunol
.
2021
;
12
:
605726
.

18

Takahashi
T
 et al.  
Higher abatacept exposure after transplant decreases acute GVHD risk without increasing adverse events
.
Blood
.
2023
;
142
:
700
710
.

19

Watkins
B
,
Williams
KM.
 
Controversies and expectations for the prevention of GVHD: A biological and clinical perspective
.
Front Immunol
.
2022
;
13
:
1057694
.

20

Lin
A
 et al.  
Letermovir for prevention of cytomegalovirus reactivation in haploidentical and mismatched adult donor allogeneic hematopoietic cell transplantation with post-transplantation cyclophosphamide for graft-versus-host disease prophylaxis
.
Biol Blood Marrow Transplant
.
2021
;
27
:85.e1–85.e6.

21

Depil
S
,
Duchateau
P
,
Grupp
SA
,
Mufti
G
,
Poirot
L.
 
‘Off-the-shelf’ allogeneic CAR T cells: development and challenges
.
Nat Rev Drug Discov
.
2020
;
19
:
185
199
.

22

Dimitri
A
,
Herbst
F
,
Fraietta
JA.
 
Engineering the next-generation of CAR T-cells with CRISPR-Cas9 gene editing
.
Mol Cancer
.
2022
;
21
:
78
.

23

Rotolo
A
 et al.  
Enhanced anti-lymphoma activity of CAR19-iNKT cells underpinned by dual CD19 and CD1d targeting
.
Cancer Cell
.
2018
;
34
:
596
610.e11
.

24

Capsomidis
A
 et al.  
Chimeric antigen receptor-engineered human gamma delta T cells: enhanced cytotoxicity with retention of cross presentation
.
Molecular Therapy
.
2018
;
26
:
354
365
.

25

Cruz
CRY
 et al.  
Infusion of donor-derived CD19-redirected virus-specific T cells for B-cell malignancies relapsed After allogeneic stem cell transplant: a phase 1 study
.
Blood
.
2013
;
122
:
2965
2973
.

26

Eyquem
J
 et al.  
Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection
.
Nature
.
2017
;
543
:
113
117
.

27

Poirot
L
 et al.  
Multiplex genome-edited T-cell manufacturing platform for “off-the-shelf” adoptive T-cell immunotherapies
.
Cancer Res
.
2015
;
75
:
3853
3864
.

28

Wang
D
,
Quan
Y
,
Yan
Q
,
Morales
JE
,
Wetsel
RA.
 
Targeted disruption of the β 2-microglobulin gene minimizes the immunogenicity of human embryonic stem cells
.
Stem Cells Transl Med
.
2015
;
4
:
1234
1245
.

29

Finke
J
,
Zeiser
R.
 
Harnessing GVHD by interleukin-6 receptor blockade
.
The Lancet Oncology
.
2014
;
15
:
1411
1412
.

30

Yalniz
FF
 et al.  
Safety and efficacy of infliximab therapy in the setting of steroid-refractory acute graft-versus-host disease
.
Biol Blood Marrow Transplant
.
2017
;
23
:
1478
1484
.

31

Bolaños-Meade
J
 et al.  
Post-transplantation cyclophosphamide-based graft-versus-host disease prophylaxis
.
N Engl J Med
 
2023
;
388
:
2338
2348
.

32

Zeiser
R
 et al.  
Ruxolitinib in corticosteroid-refractory graft-versus-host disease after allogeneic stem cell transplantation: a multicenter survey
.
Leukemia
.
2015
;
29
:
2062
2068
.

33

Algeri
M
,
Becilli
M
,
Locatelli
F.
 
Ruxolitinib as the first post-steroid treatment for acute and chronic graft-versus-host disease
.
Expert Rev Clin Immunol
 
2023
;
19
:
1299
1313
.

34

Bolaños-Meade
J
 et al.  
Phase 3 clinical trial of steroids/mycophenolate mofetil vs steroids/placebo as therapy for acute GVHD: BMT CTN 0802
.
Blood
.
2014
;
124
:
3221
3227; quiz 3335
.

35

Brown
RA
,
Byersdorfer
CA.
 
Metabolic pathways in alloreactive T cells
.
Front Immunol
 
2020
;
11
:
1517
.

36

Tijaro-Ovalle
NM
,
Karantanos
T
,
Wang
HT
,
Boussiotis
VA.
 
Metabolic targets for improvement of allogeneic hematopoietic stem cell transplantation and graft-vs.-host disease
.
Front Immunol
 
2019
;
10
:
295
.

37

Tkachev
V
 et al.  
Programmed death-1 controls T cell survival by regulating oxidative metabolism
.
J Immunol
.
2015
;
194
:
5789
5800
.

38

Mathewson
ND
 et al.  
Gut microbiome-derived metabolites modulate intestinal epithelial cell damage and mitigate graft-versus-host disease
.
Nat Immunol
.
2016
;
17
:
1235
.

39

Ramgopal
A
 et al.  
AMPK drives both glycolytic and oxidative metabolism in murine and human T cells during graft-versus-host disease
.
Blood Adv
.
2024
;
8
:
4149
4162
.

40

Huang
Y
 et al.  
Targeting glycolysis in alloreactive T cells to prevent acute graft-versus-host disease while preserving graft-versus-leukemia effect
.
Front Immunol
 
2022
;
13
:
751296
.

41

Gatza
E
 et al.  
Manipulating the bioenergetics of alloreactive T cells causes their selective apoptosis and arrests graft-versus-host disease
.
Sci Transl Med
.
2011
;
3
:
67ra8
.

42

Sena
LA
 et al.  
Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling
.
Immunity
.
2013
;
38
:
225
236
.

43

Tarasenko
TN
 et al.  
Cytochrome c oxidase activity is a metabolic checkpoint that regulates cell fate decisions during T cell activation and differentiation
.
Cell Metab
.
2017
;
25
:
1254
1268.e7
.

44

Tan
H
 et al.  
Integrative proteomics and phosphoproteomics profiling reveals dynamic signaling networks and bioenergetics pathways underlying T cell activation
.
Immunity
.
2017
;
46
:
488
503
.

45

Buck
MD
 et al.  
Mitochondrial dynamics controls T cell fate through metabolic programming
.
Cell
.
2016
;
166
:
63
76
.

46

Steinert
EM
,
Vasan
K
,
Chandel
NS.
 
Mitochondrial metabolism regulation of T cell-mediated immunity
.
Annu Rev Immunol
.
2021
;
39
:
395
416
.

47

Muehler
A
,
Peelen
E
,
Kohlhof
H
,
Gröppel
M
,
Vitt
D.
 
Vidofludimus calcium, a next generation DHODH inhibitor for the treatment of relapsing-remitting multiple sclerosis
.
Mult Scler Relat Disord
.
2020
;
43
:
102129
.

48

Malla
B
 et al.  
Teriflunomide preserves peripheral nerve mitochondria from oxidative stress-mediated alterations
.
Ther Adv Chronic Dis
.
2020
;
11
:
2040622320944773
.

49

Göttle
P
 et al.  
Teriflunomide promotes oligodendroglial differentiation and myelination
.
J Neuroinflammation
.
2018
;
15
:
76
.

50

Sykes
DB.
 
The emergence of dihydroorotate dehydrogenase (DHODH) as a therapeutic target in acute myeloid leukemia
.
Expert Opin Ther Targets
.
2018
;
22
:
893
898
.

51

Klotz
L
 et al.  
Teriflunomide treatment for multiple sclerosis modulates T cell mitochondrial respiration with affinity-dependent effects
.
Sci Transl Med
.
2019
;
11
:
490
.

52

Elgamal
OA
 et al.  
Pyrimidine depletion enhances targeted and immunotherapy combinations in 1 acute myeloid leukemia
.
JCI Insight
.
2024
;(
9
: ):

53

Cooke
KR
 et al.  
An experimental model of idiopathic pneumonia syndrome after bone marrow transplantation: 1. The roles of minor H antigens and endotoxin
.
Blood
.
1996
;
88
:
3230
3239
.

54

Neidemire-Colley
L
 et al.  
CRISPR/Cas9 deletion of MIR155HG in human T cells reduces incidence and severity of acute GVHD in a xenogeneic model
.
Blood Adv
.
2024
;
8
:.

55

Benjamini
Y
,
Krieger
AM
,
Yekutieli
D.
 
Adaptive linear step-up procedures that control the false discovery rate
.
Biometrika
.
2006
;
93
:
491
507
.

56

Nieto
Y
 et al.  
Tacrolimus and mycophenolate mofetil after nonmyeloablative matched-sibling donor allogeneic stem-cell transplantations conditioned with fludarabine and low-dose total body irradiation
.
Biol Blood Marrow Transplant
.
2006
;
12
:
217
225
.

57

Broen
JCA
,
van Laar
JM.
 
Mycophenolate mofetil, azathioprine and tacrolimus: mechanisms in rheumatology
.
Nat Rev Rheumatol
.
2020
;
16
:
167
178
. Vol,

58

Confavreux
C
 et al.  
Oral teriflunomide for patients with relapsing multiple sclerosis (TOWER): a randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet Neurol
.
2014
;
13
:
247
256
.

59

Anderson
BE
 et al.  
Memory CD4+ T cells do not induce graft-versus-host disease
.
J Clin Invest
 
2003
;
112
:
101
108
.

60

Zheng
H
,
Matte-Martone
C
,
Jain
D
,
McNiff
J
,
Shlomchik
WD.
 
Central memory CD8+ T cells induce graft-versus-host disease and mediate graft-versus-leukemia
.
J Immunol
.
2009
;
182
:
5938
5948
.

61

Scherer
S
 et al.  
Pyrimidine de novo synthesis inhibition selectively blocks effector but not memory T cell development
.
Nat Immunol
.
2023
;
24
:
501
515
.

62

Kempkes
RWM
,
Joosten
I
,
Koenen
HJPM
,
He
X.
 
Metabolic pathways involved in regulatory T cell functionality
.
Front Immunol
 
2019
;
10
:
2839
.

63

Michalek
RD
 et al.  
Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets
.
J Immunol
.
2011
;
186
:
3299
3303
.

64

Pearce
EL.
 
Metabolism in T cell activation and differentiation
.
Curr Opin Immunol
.
2010
;
22
:
314
320
.

65

Li
Q
 et al.  
Donor T cell STAT3 deficiency enables tissue PD-L1–dependent prevention of graft-versus-host disease while preserving graft-versus-leukemia activity
.
J Clin Invest
.
2023
;
133
:

66

Bleakley
M
 et al.  
Naive T-cell depletion to prevent chronic graft-versus-host disease
.
JCO
 
2022
;
40
:
1174
1185
.

Author notes

Rathan Kumar and Kara M. Braunreiter contributed equally.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)

Supplementary data