Abstract
Purpose of Review:
Controlling T cell activity through metabolic manipulation has become a prominent feature in immunology and practitioners of both adoptive cellular therapy (ACT) and hematopoietic stem cell transplantation (HSCT) have utilized metabolic interventions to control T cell function. This review will survey recent metabolic research efforts in HSCT and ACT to paint a broad picture of immunometabolism and highlight advances in each area.
Recent Findings:
In HSCT, recent publications have focused on modifying reactive oxygen species, sirtuin signaling, or the NAD salvage pathway within alloreactive T cells, and to a lesser extent on modulating regulatory T cells. In ACT, metabolic interventions that bolster memory T cell development, increase mitochondrial density and function, or block regulatory signals in the tumor microenvironment (TME) have recently been published.
Summary:
Metabolic interventions control immune responses. In ACT, efforts seek to improve the in vivo metabolic fitness of T cells, while in HSCT energies have focused on blocking alloreactive T cell expansion or promoting regulatory T cells. Methods to identify new, metabolically targetable pathways, as well as the ability of metabolic biomarkers to predict disease onset and therapeutic response, will continue to advance the field towards clinically applicable interventions.
Keywords: Immunometabolism, hematopoietic stem cell transplantation, adoptive cellular therapy (ACT), CAR-T cells, tumor-infiltrating lymphocytes (TIL)
Introduction
Controlling T cell activity has become the Holy Grail of clinical immunology and reliance on oxidative versus glycolytic metabolism exerts significant influence over T cell fate. However, to make metabolic control a clinical reality, it is crucial to understand metabolism within specific environmental contexts. We will begin by reviewing the influence of metabolism on T cell activation and differentiation, then cover recent advances in understanding T cell metabolism during graft-versus-host disease (GVHD), metabolism-based methods to improve adoptive cellular therapies (ACT), and a role for metabolic biomarkers following hematopoietic stem cell transplantation (HSCT).
The role of T cell metabolism in differentiation and activation
In broad strokes, T cell metabolism can be divided into oxidative phosphorylation (OXPHOS), which uses the proton gradient established by the electron transport chain (ETC) to generate ATP, and glycolysis, where glucose is converted into lactate in the absence of oxidation. OXPHOS generates ATP more efficiently, but glycolysis supports one carbon metabolism, a necessity during rapid T cell expansion[1]. Naïve and memory cells are metabolically quiescent, while effector T cells (Teff) aggressively increase their metabolism to support proliferation and effector functions. In the classic paradigm, naïve and memory T cells adopt an oxidative phenotype, while Teff rely more heavily on glycolysis[2]. Regulatory T cells (Treg) adopt their own unique metabolic signature[3]. However, these categories were largely established in vitro and it is increasingly clear that in vitro metabolic pathways do not necessarily reflect those utilized by T cells in vivo[4,5], with ample evidence that Teff can adopt an oxidative phenotype[6–8]. In allogeneic HSCT, the intent is to inhibit Teff while promoting Treg, while the goal for ACT is to do the opposite. Despite these opposing objectives, common ways to understand and manipulate T cell metabolism link these fields together and much can be learned from understanding the techniques and targets employed by the other side.
Cellular Metabolism in GVHD
The challenge facing T cell manipulation during allogeneic HSCT is how to mitigate GVHD-causing T cells while still preserving graft-versus-tumor (GVT) responses. One way to achieve this goal is by improving the Treg/Teff ratio, either by decreasing allogeneic Teff numbers or increasing the number or stability of the Treg population[9–11].
Metabolic control of alloreactive T cells
Reactive oxygen species (ROS) propagate several aspects of T cell-mediated immunity including intracellular signaling downstream of the T cell receptor (TCR)[12,13]. High levels of ROS were found in the liver, spleen, and bone marrow during acute GVHD [14,15] and promotion of ROS scavenging, through T cell overexpression of thioredoxin-1 (Trx1), reduced GVHD development [14]. Systemically limiting ROS with apocynin, a pharmacologic NADPH oxidase inhibitor, similarly reduced GVHD clinical scores and improved recipient survival [15]. Interestingly, Trx1 overexpression decreased glutamine uptake in liver T cells, but not those recovered from the spleen [14], suggesting metabolic reliance based in part on cellular location. Intracellular ROS levels are controlled by redox pathways, and the transcription factor Nuclear factor erythroid 2–related factor 2 (Nrf2) controls expression of an array of antioxidant genes. Transplantation of Nrf2-deficient T cells reduced GVHD clinical scores and improved post-transplant survival [16]. Interestingly, systemic treatment with arsenic trioxide, which upregulates NRF2 globally, minimized GVHD pathogenesis [17], highlighting the need for further study of Nrf2’s role in the post-transplant environment.
Sirtuins are class III histone deacetylases (HDACs) and Sirtuin 1 (SIRT1) controls cellular metabolism by deacetylating a broad range of transcription factors and transcriptional co-regulators. SIRT3 is a mitochondrially-associated HDAC which inhibits intracellular ROS generation. Transplantation of SIRT1-deficient T cells decreased GVHD by limiting CD4+ cell division and minimizing interferon gamma (IFN-γ) production. Transplantation of SIRT3-deficient T cells also decreased GVHD severity and improved post-transplant mortality [18], but ROS levels actually decreased in these cells, a likely consequence of reduced T cell activation. Sirtuins and T cells both require nicotinamide (NAD) as a co-factor for their function, making them dependent upon the NAD salvage pathway, a process catalyzed by nicotinamide phosphoribosyl-transferase (NAMPT) and intricately linked to glucose metabolism [19]. Serum NAMPT levels increased in acute GVHD models and pharmacologic blockade of NAMPT limited liver- and spleen-associated donor CD4+ and CD8+ T cell numbers, with improved clinical scores and reduced post-transplant weight loss [20]. Interestingly, NAMPT inhibition exerted anti-leukemia effects independent of its impact on GVHD T cells, while SIRT1, SIRT3, and NRF2-deficient donor T cells mitigated GVHD while still preserving GVT responses [18,21].
Metabolic augmentation of Treg
Both genetic and pharmacologic downregulation of ROS favored FoxP3 expression, the transcription factor synonymous with Treg generation. T cell overexpression of Trx1 increased Foxp3 levels in recipient spleens [14] and recipients of Nrf2 knock-out (KO) cells had greater Treg percentages with increased expression of Helios, a marker of thymus-derived Tregs [16]. Inhibition of the NAD salvage pathway also improved Treg/Teff ratios [20] and transplantation of SIRT1 KO T cells increased Foxp3 percentages, with fewer Treg converting into INF-γ producing Th1 cells [21]. A caveat to interpreting studies where the gene is knocked out in all T cells is whether the metabolic intervention directly impacts Tregs themselves, or simply lessens systemic inflammation, which improves Treg stability.
Treg administration can improve the post-transplant Treg/Teff balance and adoptive transfer of Treg lacking vimentin mitigated GVHD to a greater degree than WT Treg, a consequence of enhanced oxidative metabolism and concomitant increase in suppressive capacity [22]. Infusion of CD4CD8 double negative (DN) Treg cells also prevented xenogeneic GVHD [23] and levels of DN Treg correlated with chronic GVHD (cGVHD) severity [24]. Teff co-cultured with DN Treg decreased mammalian target of rapamycin (mTOR) signaling [25], but whether this decrease was simply a consequence of reduced Teff proliferation is unclear.
Metabolic control in non-T cells
Retinoic acid (RA), a metabolite of Vitamin A, is critical for development of committed CD4 and CD8 T cells and during GVHD, RA is pro-inflammatory and high levels correlate with more severe disease. A lack of RA synthesis in host DCs impaired Teff function, increased donor Treg frequency, and improved survival [26]. These changes were not seen if intestinal epithelial cells (IECs) lacked RA synthesis, suggesting that RA metabolism in host DCs (> donor DCs) is critical to regulate Treg/Teff ratios. Previous studies have also described salutary effects of butyrate on intestinal epithelial cells (IECs) [27]. Expression of the G-protein coupled receptor GPR43 was necessary for butyrate’s protective effects and epithelial repair was secondary to secretion of local cytokines and subsequent activation of the inflammasome [28].
Metabolic challenges in adoptive cellular therapy
Repurposing a patient’s own immune system to combat cancer is a unique form of ACT and includes expansion of tumor infiltrating lymphocytes (TIL) and creation of chimeric antigen receptor (CAR) T cells. While these methods seek to reinvigorate underperforming immune responses, they face many metabolic challenges. T cells obtained from tumor sites or the peripheral blood are often terminally differentiated or “exhausted” and require significant activation in vitro. Unfortunately, this stimulation drives terminal differentiation, leading to poor activity and limited persistence upon return to the host [29]. After transfer, T cells need to survive the harsh metabolic landscape of the in vivo environment, made worse in fighting leukemia because leukemic cells block T cell acquisition of glucose [30]. Furthermore, there is a clear correlation between loss of cancer-targeting T cells and relapse [31]. Thus, techniques to support or encourage the metabolic health of tumor-specific T cells are critical.
Memory T cells (Tmem) adopt a phenotype skewed towards oxidative metabolism and encouraging T cells to become metabolically more “memory-like” gives them a critical adaptation upon in vivo transfer. For example, CAR T cells bearing the CD28 co-stimulatory domain adopt a glycolysis-driven phenotype, with aggressive early activation followed by poor in vivo persistence. In contrast, CAR T cells bearing the 4–1BB co-receptor relied on oxidative metabolism, with a slower early expansion but improved overall persistence [32]. Promoting a more oxidative phenotype can be addressed in two main ways. The first utilizes metabolic inhibitors, nutrient withdrawal/supplementation, cytokine addition, and checkpoint blockade to alter the environment in which T cells expand or ultimately function. The second approach genetically modifies T cells to delete unwanted mediators, block ineffective metabolic signaling, or promote advantageous metabolic pathways.
External Manipulation: Targeting the T cell Environment
T cell expansion is classically glycolysis-driven. Thus, growing T cells in 2-deoxyglucose (2DG), an inhibitor of hexokinase, increased the percentage of memory-like cells and improved their tumor clearance in a mouse model of melanoma[33]. In addition, the IL-2 used during in vitro expansion induces glycolysis by upregulating expression of lactate dehydrogenase (LDH), the terminal step in glycolysis. Expansion in IL-21 reduced LDH induction and increased shuttling of pyruvate into the mitochondria. Simultaneous pharmacological blockade of LDH enhanced memory formation with reduced expression of exhaustion markers.[34] Glutamine blockade similarly restricted T cell differentiation and improved anti-tumor function post-transfer, with enhanced generation of memory cells[35]. ROS levels also impacted in vitro differentiation of ACT T cells. Addition of the anti-oxidant N-acetylcysteine during expansion produced less-differentiated stem and central memory cells which were more functional [36,37], while ROS induction conversely increased the percentage of more differentiated effector memory cells.
Targeting T cells in the TME is another potential opportunity to metabolically augment T cell activity Targeting programmed death 1 (PD-1) improves T cell function in the TME[38]. Recently, it has become clear that PD-1 signaling is intricately linked to cellular metabolism. The majority of transcripts altered by PD-1 ligation involved metabolic pathways, with blunting of both glycolysis and oxidative metabolism[39]. In other work, T cells expressing an oncolytic vaccinia virus increased influx of T cells into the TME, however these cells showed poor activity and decreased mitochondrial reserve. This metabolic dysfunction was reversed through enhanced leptin signaling as leptin delivered to the TME via vaccinia virus expression improved T cell mitochondrial density and enhanced spare respiratory capacity (SRC), ultimately resulting in more efficient tumor clearance[40]. Other factors, including the balance between CD4+ and CD8+ T cells, influence CD8+ T cell subset formation and function. These factors could also improve the effectiveness of ACT. Recent evidence from a viral model suggests that CD8+ T cells generated with adequate CD4+ T cell help enhance their cytolytic capacity, SRC, and proliferation upon secondary activation exposure compared to cells without adequate CD4+ help [41]. Thus, improving CD4+ T cell yields during in vitro expansion may be the best guarantee of functional CD8 T cells in vivo.
Internal Manipulation: Genetically Modifying T cells
Blockade of specific metabolic pathways can be done externally as discussed above, but a more permanent way to alter T metabolic choice is through genetic manipulation. For example, T cells can be modified to ignore messages of metabolic dysfunction transmitted by transforming growth factor-beta (TGF-β), which in the TME blocks both T cell glycolysis and oxidative metabolism [42]. As expected, deletion of the TGF-β receptor in adoptively transferred cells improved overall anti-tumor activity.[43] Alternatively, oxidative proteins can be overexpressed to promote advantageous metabolism. Targets for this endeavor include the mitochondrial fusion protein Opa1 and those controlling mitochondrial biogenesis, like peroxisome proliferator activated receptor co-activator 1-alpha (PGC1α). These metabolic interventions are modeled after characteristic changes in Tmem mitochondrial density and organization and Opa1 overexpression in ACT generated more memory-like T cells with improved in vivo efficacy [44]. Similarly, increased expression of PGC1α enhanced central memory T cell formation with characteristic increased SRC and augmented proliferation upon antigen re-challenge[45]. In addition to these well-described targets, additional methods to screen for metabolically influential proteins have been developed. Using a CRISPR/Cas9 library screen to knock down candidate genes, REGNASE-1 was identified as playing a key role in metabolic exhaustion. Previously known as an RNAse with RNA binding activity, REGNASE-1 depleted T cells showed enhanced anti-tumor activity[46]. This finding synergized with simultaneous knockdown of additional proteins from the same screen, indicating that further targets may continue to be identified through this method.
Metabolism-based biomarkers
While metabolic manipulation has shown promising results in GVHD and ACT, identifying patients who would benefit most from specific interventions remains an area of active investigation. Allogeneic transplantation in humans is followed by major changes in recipient serum metabolic profiles through a combination of microbial dysbiosis [47] and transplantation-related alteration of host metabolism. Patient samples experienced a significant decrease in tryptophan metabolites at the onset of acute GvHD, including a large decrease in microbiota-produced compounds such as 3-indoxyl sulfate (3-IS) [48]. Previous studies correlated low urinary 3-IS levels with GVHD severity, while increased indole intake improved GVHD-associated weight loss and extended post-transplant survival [49]. Dietary intake of choline has an effect opposite of indoles and oral administration of the choline derivative Tri-methylamine N-oxide (TMAO) worsened GVHD, an effect driven by inflammasome activation, enhanced M1 macrophage polarization, and production of IL-1β [50]. Conversely, treatment of allogeneic recipients with arsenic trioxide decreased macrophage M1 polarization and improved GVHD [51]. These data suggest that reducing choline-mediated macrophage inflammation, while providing protective indoles or short-chain fatty acids, could synergistically protect against GVHD.
Amino acids levels also have predictive value post-transplant. Metabolomic analysis on day 0 (prior to HSCT infusion) identified 20 metabolites associated with GVHD [52], among them a decrease in 2-aminobutyric acid. In addition, total plasma amino acids decreased on day +7 in patients who later developed severe acute GVHD [53], while decreased levels of the branched chain amino acids leucine and isoleucine on day 100 predicted cGVHD development [54]. It is hypothesized that amino acids are consumed as tissues are damaged and immune cell activation programs (including cytokine production) are initiated. In contrast to amino acids, complex lipid products including medium- and long-chain fatty acid (FA), polyunsaturated FA, diacylglycerol, and primary/secondary bile acids increased in patients destined to develop or already experiencing acute GvHD [48,52]. FA levels also increased in future cGVHD patients [54]. A separate approach combining lipidomics and transcriptomics revealed a robust increase in glycerophospholipid (GPL) metabolism and allowed construction of an aGvHD risk score with both diagnostic and prognostic value [55]. Together these data suggest that serum metabolic profiles can function as indirect indicators of T cell activation and thus serve to both gauge GVHD as well as evaluate responses to therapeutic interventions.
The use of biomarkers to predict success in ACT has focused mainly on expression of inhibitory ligands in the TME and serum cytokine production. Monitoring tumors for PDL1 expression is a potential marker for the utility of checkpoint inhibition, although this approach does not guarantee successful therapy [56]. Similarly, measurement of inflammatory cytokines such as IL-6 have been used to indicate CAR-T cell activity [57]. Assessments of true metabolic intermediates may help shed light, as it has in GVHD, on the metabolic activity of tumors and suggest therapies to best target them.
Conclusion
The study of T cell metabolism has opened many doors for understanding and controlling T cell immunity. During GVHD, defining metabolic pathways in Treg and alloreactive Teff will allow for GHVD-specific interventions with preservation of physiologic and graft-versus-tumor responses. In ACT, recognizing the metabolic dysfunction caused by in vitro T cell expansion and the TME can direct interventions to specifically augment metabolic health during key times to improve anti-tumor efficacy. Despite promising preclinical work, the clinical utility of many interventions continues to fall short. Thus, research focusing on predictive biomarkers may identify patients at risk for GVHD, those who will benefit most from specific interventions (e.g. checkpoint inhibitor therapy), or better predict treatment efficacy in both HSCT and ACT. Overall, our ongoing gains in understanding T cell metabolism will increase the ability to manipulate metabolic pathways to clinical advantage and deliver exciting new therapies to patients who are most at-risk.
Key Points.
In HSCT, modulation of ROS levels, sirtuin signaling and the NAD salvage pathway were used to decrease Teff and increased Treg post-transplant.
In ACT, augmentation of oxidative metabolism and a decrease in inhibitory signaling (e.g. TGF-β) overcame metabolic dysfunction caused by in vitro expansion and influences of the TME
Metabolic biomarkers could be used to predict the onset of GVHD as well as the potential response to therapy in both HSCT and ACT.
Acknowledgements
This work was supported by grants to CB from the National Institute of Health—NHBLI (K08 HL123631, R01 HL144556), the Department of Defense (CA180681), the Hyundai Motor Company (Hope on Wheels Scholar grant), the American Society of Hematology (Scholar award), and the Be the Match Foundation (Amy Strelzer Manasevit award) and to EB from the Burroughs Wellcome Fund (Physician Scientist Incubator program), the NCI (CA082084), the NIH (K12 HD052892), and the St. Baldrick’s Foundation (Fellowship award).
Footnotes
Conflict of Interest
None
Bibliography
- 1.Lunt SY, Vander Heiden MG: Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol 2011, 27:441–464. [DOI] [PubMed] [Google Scholar]
- 2.Geltink RIK, Kyle RL, Pearce EL: Unraveling the complex interplay between T cell metabolism and function. Annu. Rev. Immunol 2018, 36:461–488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Coe DJ, Kishore M, Marelli-Berg F: Metabolic regulation of regulatory T cell development and function. Front. Immunol 2014, 5:590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.**.Ma EH, Verway MJ, Johnson RM, Roy DG, Steadman M, Hayes S, Williams KS, Sheldon RD, Samborska B, Kosinski PA, et al. : Metabolic profiling using stable isotope tracing reveals distinct patterns of glucose utilization by physiologically activated CD8+ T cells. Immunity 2019, 51:856–870.e5. [DOI] [PubMed] [Google Scholar]; This paper supports the long-held belief that in vitro T cell metabolism does not necessarily reflect in vivo metabolic adaptations and highlighted an increased role for oxidative metabolism in vivo in recently activated CD8 T cells.
- 5.Allocco JB, Alegre M-L: In vivo and in vitro differences in CD8+ T cell metabolism. Am. J. Transplant 2020, 20:327. [Google Scholar]
- 6.Blagih J, Coulombe F, Vincent EE, Dupuy F, Galicia-Vázquez G, Yurchenko E, Raissi TC, van der Windt GJW, Viollet B, Pearce EL, et al. : The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity 2015, 42:41–54. [DOI] [PubMed] [Google Scholar]
- 7.Gatza E, Wahl DR, Opipari AW, Sundberg TB, Reddy P, Liu C, Glick GD, Ferrara JLM: Manipulating the bioenergetics of alloreactive T cells causes their selective apoptosis and arrests graft-versus-host disease. Sci. Transl. Med 2011, 3:67ra8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Byersdorfer CA, Tkachev V, Opipari AW, Goodell S, Swanson J, Sandquist S, Glick GD, Ferrara JLM: Effector T cells require fatty acid metabolism during murine graft-versus-host disease. Blood 2013, 122:3230–3237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Edinger M, Hoffmann P, Ermann J, Drago K, Fathman CG, Strober S, Negrin RS: CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat. Med 2003, 9:1144–1150. [DOI] [PubMed] [Google Scholar]
- 10.*.Hippen KL, Aguilar EG, Rhee SY, Bolivar-Wagers S, Blazar BR: Distinct Regulatory and Effector T Cell Metabolic Demands during Graft-Versus-Host Disease. Trends Immunol. 2020, 41:77–91. [DOI] [PMC free article] [PubMed] [Google Scholar]; A wonderful review on the distinct metabolic adaptations between Teff and Treg cells during GVHD.
- 11.Hippen KL, Merkel SC, Schirm DK, Sieben CM, Sumstad D, Kadidlo DM, McKenna DH, Bromberg JS, Levine BL, Riley JL, et al. : Massive ex vivo expansion of human natural regulatory T cells (T(regs)) with minimal loss of in vivo functional activity. Sci. Transl. Med 2011, 3:83ra41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Rashida Gnanaprakasam JN, Wu R, Wang R: Metabolic reprogramming in modulating T cell reactive oxygen species generation and antioxidant capacity. Front. Immunol 2018, 9:1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Franchina DG, Dostert C, Brenner D: Reactive oxygen species: involvement in T cell signaling and metabolism. Trends Immunol. 2018, 39:489–502. [DOI] [PubMed] [Google Scholar]
- 14.*.Sofi MH, Wu Y, Schutt SD, Dai M, Daenthanasanmak A, Heinrichs Voss J, Nguyen H, Bastian D, Iamsawat S, Selvam SP, et al. : Thioredoxin-1 confines T cell alloresponse and pathogenicity in graft-versus-host disease. J. Clin. Invest 2019, 129:2760–2774. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this article, authors use overexpression of Thioredoxin-1 to investigate the role of ROS in the development of GVHD.
- 15.Rezende BM, Bernardes PTT, Gonçalves WA, de Resende CB, Athayde RM, Ávila TV, Martins DG, Castor MGM, Teixeira MM, Pinho V: Treatment with Apocynin Limits the Development of Acute Graft-versus-Host Disease in Mice. J. Immunol. Res 2019, 2019:9015292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Tsai JJ, Velardi E, Shono Y, Argyropoulos KV, Holland AM, Smith OM, Yim NL, Rao UK, Kreines FM, Lieberman SR, et al. : Nrf2 regulates CD4+ T cell-induced acute graft-versus-host disease in mice. Blood 2018, 132:2763–2774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hu X, Li L, Yan S, Li Z: Arsenic trioxide suppresses acute graft-versus-host disease by activating the Nrf2/HO-1 pathway in mice. Br. J. Haematol 2019, 186:e145–e148. [DOI] [PubMed] [Google Scholar]
- 18.Toubai T, Tamaki H, Peltier DC, Rossi C, Oravecz-Wilson K, Liu C, Zajac C, Wu J, Sun Y, Fujiwara H, et al. : Mitochondrial Deacetylase SIRT3 Plays an Important Role in Donor T Cell Responses after Experimental Allogeneic Hematopoietic Transplantation. J. Immunol 2018, 201:3443–3455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Murphy JP, Giacomantonio MA, Paulo JA, Everley RA, Kennedy BE, Pathak GP, Clements DR, Kim Y, Dai C, Sharif T, et al. : The NAD+ Salvage Pathway Supports PHGDH-Driven Serine Biosynthesis. Cell Rep. 2018, 24:2381–2391.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.**.Gerner RR, Macheiner S, Reider S, Siegmund K, Grabherr F, Mayr L, Texler B, Moser P, Effenberger M, Schwaighofer H, et al. : Targeting NAD immunometabolism limits severe graft-versus-host disease and has potent antileukemic activity. Leukemia 2020, doi: 10.1038/s41375-020-0709-0. [DOI] [PubMed] [Google Scholar]; In this article authors note a clear increase in serum NAMPT levels in GVHD patients post-transplant and make a compelling case for the role of the NAD salvage pathway in the pathogenesis of GVHD. In addition, they demonstrate that inhibition of NAD metabolism has anti-leukemia properties independent of its effects on T cells.
- 21.**.Daenthanasanmak A, Iamsawat S, Chakraborty P, Nguyen HD, Bastian D, Liu C, Mehrotra S, Yu X-Z: Targeting Sirt-1 controls GVHD by inhibiting T-cell allo-response and promoting Treg stability in mice. Blood 2019, 133:266–279. [DOI] [PMC free article] [PubMed] [Google Scholar]; Another solid paper demonstrating a role for Sirt1 in both CD4+ Teff as well as in the Treg following allogeneic transplantation in mice.
- 22.**.McDonald-Hyman C, Muller JT, Loschi M, Thangavelu G, Saha A, Kumari S, Reichenbach DK, Smith MJ, Zhang G, Koehn BH, et al. : The vimentin intermediate filament network restrains regulatory T cell suppression of graft-versus-host disease. J. Clin. Invest 2018, 128:4604–4621. [DOI] [PMC free article] [PubMed] [Google Scholar]; Authors use a phosphoproteomic screen to identify the intermediate filament vimentin as a target of PKC-θ. In addition, disruption of vimentin by siRNA or treatment with a clinically relevant PKC-θ inhibitor enhanced Treg metabolic and suppressive activity compared to wildtype cells.
- 23.Achita P, Dervovic D, Ly D, Lee JB, Haug T, Joe B, Hirano N, Zhang L: Infusion of ex-vivo expanded human TCR-αβ+ double-negative regulatory T cells delays onset of xenogeneic graft-versus-host disease. Clin. Exp. Immunol 2018, 193:386–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hillhouse EE, Thiant S, Moutuou MM, Lombard-Vadnais F, Parat R, Delisle J-S, Ahmad I, Roy D-C, Guimond M, Roy J, et al. : Double-Negative T Cell Levels Correlate with Chronic Graft-versus-Host Disease Severity. Biol. Blood Marrow Transplant 2019, 25:19–25. [DOI] [PubMed] [Google Scholar]
- 25.*.Haug T, Aigner M, Peuser MM, Strobl CD, Hildner K, Mougiakakos D, Bruns H, Mackensen A, Völkl S: Human Double-Negative Regulatory T-Cells Induce a Metabolic and Functional Switch in Effector T-Cells by Suppressing mTOR Activity. Front. Immunol 2019, 10:883. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this manuscript, co-culture of human effector T cells with CD4/CD8 double-negative regulatory T cells decreased proliferation of the Teff population along with a marked decrease in mTOR signaling.
- 26.*.Thangavelu G, Lee Y-C, Loschi M, Schaechter KM, Feser CJ, Koehn BH, Nowak EC, Zeiser R, Serody JS, Murphy WJ, et al. : Dendritic Cell Expression of Retinal Aldehyde Dehydrogenase-2 Controls Graft-versus-Host Disease Lethality. J. Immunol 2019, 202:2795–2805. [DOI] [PMC free article] [PubMed] [Google Scholar]; Previous work has shown that retinoic acid is inflammatory during GVHD, while decreased RA signaling in T cells mitigated GVHD pathogenesis. In this manuscript, authors demonstrate that conditional deletion of the RA-synthesizing enzyme RALDH2 in host dendritic cells reduces GVHD lethality.
- 27.Mathewson ND, Jenq R, Mathew AV, Koenigsknecht M, Hanash A, Toubai T, Oravecz-Wilson K, Wu S-R, Sun Y, Rossi C, et al. : Gut microbiome-derived metabolites modulate intestinal epithelial cell damage and mitigate graft-versus-host disease. Nat. Immunol 2016, 17:505–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.*.Fujiwara H, Docampo MD, Riwes M, Peltier D, Toubai T, Henig I, Wu SJ, Kim S, Taylor A, Brabbs S, et al. : Microbial metabolite sensor GPR43 controls severity of experimental GVHD. Nat. Commun 2018, 9:3674. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this manuscript, authors demonstrated that the metabolite sensor G-protein-coupled receptor 43 (GPR43) is critical for the protective effects of short-chain fatty acids (e.g. butyrate) and make the novel observation that GPR43 works through inflammasome activation in non-hematopoietic host tissues.
- 29.Ando M, Ito M, Srirat T, Kondo T, Yoshimura A: Memory T cell, exhaustion, and tumor immunity. Immunol. Med 2020, 43:1–9. [DOI] [PubMed] [Google Scholar]
- 30.Siska PJ, van der Windt GJW, Kishton RJ, Cohen S, Eisner W, MacIver NJ, Kater AP, Weinberg JB, Rathmell JC: Suppression of glut1 and glucose metabolism by decreased akt/mtorc1 signaling drives T cell impairment in B cell leukemia. J. Immunol 2016, 197:2532–2540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Singh N, Frey NV, Grupp SA, Maude SL: CAR T cell therapy in acute lymphoblastic leukemia and potential for chronic lymphocytic leukemia. Curr. Treat. Options Oncol 2016, 17:28. [DOI] [PubMed] [Google Scholar]
- 32.Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, et al. : Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med 2014, 371:1507–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sukumar M, Liu J, Ji Y, Subramanian M, Crompton JG, Yu Z, Roychoudhuri R, Palmer DC, Muranski P, Karoly ED, et al. : Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J. Clin. Invest 2013, 123:4479–4488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.*.Hermans D, Gautam S, García-Cañaveras JC, Gromer D, Mitra S, Spolski R, Li P, Christensen S, Nguyen R, Lin J-X, et al. : Lactate dehydrogenase inhibition synergizes with IL-21 to promote CD8+ T cell stemness and antitumor immunity. Proc. Natl. Acad. Sci. USA 2020, 117:6047–6055. [DOI] [PMC free article] [PubMed] [Google Scholar]; This manuscript reveals a significant difference in metabolic programming between IL2 and IL21 stimulated cells.
- 35.Nabe S, Yamada T, Suzuki J, Toriyama K, Yasuoka T, Kuwahara M, Shiraishi A, Takenaka K, Yasukawa M, Yamashita M: Reinforce the antitumor activity of CD8+ T cells via glutamine restriction. Cancer Sci. 2018, 109:3737–3750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.*.Pilipow K, Scamardella E, Puccio S, Gautam S, De Paoli F, Mazza EM, De Simone G, Polletti S, Buccilli M, Zanon V, et al. : Antioxidant metabolism regulates CD8+ T memory stem cell formation and antitumor immunity. JCI Insight 2018, 3. [DOI] [PMC free article] [PubMed] [Google Scholar]; This group trialed a variety of antioxidants during T cell expansion, eventually finding that NAC addition allowed for the production of less differentiated cells with enhanced anti-tumor activity after transfer.
- 37.Pilipow K, Scamardella E, Lugli E: Generating stem-like memory T cells with antioxidants for adoptive cell transfer immunotherapy of cancer. Meth. Enzymol 2020, 631:137–158. [DOI] [PubMed] [Google Scholar]
- 38.Bardhan K, Anagnostou T, Boussiotis VA: The PD1:PD-L1/2 Pathway from Discovery to Clinical Implementation. Front. Immunol 2016, 7:550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.**.Ogando J, Sáez ME, Santos J, Nuevo-Tapioles C, Gut M, Esteve-Codina A, Heath S, González-Pérez A, Cuezva JM, Lacalle RA, et al. : PD-1 signaling affects cristae morphology and leads to mitochondrial dysfunction in human CD8+ T lymphocytes. J. Immunother. Cancer 2019, 7:151. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper utilized co-signaling through the PD1 receptor during in vitro activation to highlight pathways involved in the PD1 signaling cascade.
- 40.**.Rivadeneira DB, DePeaux K, Wang Y, Kulkarni A, Tabib T, Menk AV, Sampath P, Lafyatis R, Ferris RL, Sarkar SN, et al. : Oncolytic Viruses Engineered to Enforce Leptin Expression Reprogram Tumor-Infiltrating T Cell Metabolism and Promote Tumor Clearance. Immunity 2019, 51:548–560.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this manuscript, the authors coupled immunogenic vaccinia therapy with metabolic manipulation through enhanced leptin secretion, leading to more effective tumor clearance.
- 41.**.Cullen JG, McQuilten HA, Quinn KM, Olshansky M, Russ BE, Morey A, Wei S, Prier JE, La Gruta NL, Doherty PC, et al. : CD4+ T help promotes influenza virus-specific CD8+ T cell memory by limiting metabolic dysfunction. Proc. Natl. Acad. Sci. USA 2019, 116:4481–4488. [DOI] [PMC free article] [PubMed] [Google Scholar]; This manuscript utilized a viral model to show that metabolically, CD8+ T cells with adequate CD4+ T cell help significantly increased their spare respiratory capacity.
- 42.Cattley RT, Lee M, Boggess WC, Hawse WF: Transforming growth factor β (TGF-β) receptor signaling regulates kinase networks and phosphatidylinositol metabolism during T-cell activation. J. Biol. Chem 2020, 295:8236–8251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.**.Tang N, Cheng C, Zhang X, Qiao M, Li N, Mu W, Wei X-F, Han W, Wang H: TGF-β inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors. JCI Insight 2020, [no volume]. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this manuscript, authors deleted the TGFβ receptor from CAR-T cells and were able to not only improve tumor clearance, but also enhance secondary recall responses.
- 44.Buck MD, O’Sullivan D, Klein Geltink RI, Curtis JD, Chang C-H, Sanin DE, Qiu J, Kretz O, Braas D, van der Windt GJW, et al. : Mitochondrial Dynamics Controls T Cell Fate through Metabolic Programming. Cell 2016, 166:63–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.**.Dumauthioz N, Tschumi B, Wenes M, Marti B, Wang H, Franco F, Li W, Lopez-Mejia IC, Fajas L, Ho P-C, et al. : Enforced PGC-1α expression promotes CD8 T cell fitness, memory formation and antitumor immunity. Cell Mol Immunol 2020, doi: 10.1038/s41423-020-0365-3. [DOI] [PMC free article] [PubMed] [Google Scholar]; Here authors found that overexpression of PGC1α enforced greater mitochondrial biogenesis and respiration without increasing cellular ROS.
- 46.**.Wei J, Long L, Zheng W, Dhungana Y, Lim SA, Guy C, Wang Y, Wang Y-D, Qian C, Xu B, et al. : Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy. Nature 2019, 576:471–476. [DOI] [PMC free article] [PubMed] [Google Scholar]; Utilizing a CRISPR/cas9 screening approach, authors were able to identify a key molecule which regulates anti-tumor immune responses in an unbiased way.
- 47.Wilmanski T, Rappaport N, Earls JC, Magis AT, Manor O, Lovejoy J, Omenn GS, Hood L, Gibbons SM, Price ND: Blood metabolome predicts gut microbiome α-diversity in humans. Nat. Biotechnol 2019, 37:1217–1228. [DOI] [PubMed] [Google Scholar]
- 48.**.Michonneau D, Latis E, Curis E, Dubouchet L, Ramamoorthy S, Ingram B, de Latour RP, Robin M, de Fontbrune FS, Chevret S, et al. : Metabolomics analysis of human acute graft-versus-host disease reveals changes in host and microbiota-derived metabolites. Nat. Commun 2019, 10:5695. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this elegant study, authors used metabolomic analysis of patient samples and donor controls to determine serum metabolites that derived from the transplantation procedure as well as those that were predictive of GVHD.
- 49.*.Swimm A, Giver CR, DeFilipp Z, Rangaraju S, Sharma A, Ulezko Antonova A, Sonowal R, Capaldo C, Powell D, Qayed M, et al. : Indoles derived from intestinal microbiota act via type I interferon signaling to limit graft-versus-host disease. Blood 2018, 132:2506–2519. [DOI] [PMC free article] [PubMed] [Google Scholar]; This publication demonstrates the ability of indole administration to reduce gut damage, reduce transepithelial bacterial translocation, and minimize GVHD mortality in a process that was dependent upon a type I interferon (IFN1) response in host cells.
- 50.**.Wu K, Yuan Y, Yu H, Dai X, Wang S, Sun Z, Wang F, Fei H, Lin Q, Jiang H, et al. : Gut microbial metabolite trimethylamine N-oxide aggravates GVHD by inducing M1 macrophage polarization in mice. Blood 2020, doi: 10.1182/blood.2019003990. [DOI] [PMC free article] [PubMed] [Google Scholar]; Authors demonstrate that administration of the choline metabolite trimethylamine N-oxide worsens GVHD through polarization of macrophages to an M1 phenotype and that in the absence inflammasome signaling, these changes do not occur.
- 51.Liu X, Su Y, Sun X, Fu H, Huang Q, Chen Q, Mo X, Lv M, Kong Y, Xu L, et al. : Arsenic trioxide alleviates acute graft-versus-host disease by modulating macrophage polarization. Sci China Life Sci 2020, doi: 10.1007/s11427-019-1691-x. [DOI] [PubMed] [Google Scholar]
- 52.**.Contaifer D, Roberts CH, Kumar NG, Natarajan R, Fisher BJ, Leslie K, Reed J, Toor AA, Wijesinghe DS: A Preliminary Investigation towards the Risk Stratification of Allogeneic Stem Cell Recipients with Respect to the Potential for Development of GVHD via Their Pre-Transplant Plasma Lipid and Metabolic Signature. Cancers (Basel) 2019, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]; This innovative study looked at the lipidomic and metabolomic signature in 14 patients at day 0 and from this array developed a 5-biomarker panel with a high sensitivity and specificity towards predicting post-transplant GVHD.
- 53.Weischendorff S, Kielsen K, Nederby M, Schmidt L, Burrin D, Heilmann C, Ifversen M, Sengeløv H, Mølgaard C, Müller K: Reduced Plasma Amino Acid Levels During Allogeneic Hematopoietic Stem Cell Transplantation Are Associated with Systemic Inflammation and Treatment-Related Complications. Biol. Blood Marrow Transplant 2019, 25:1432–1440. [DOI] [PubMed] [Google Scholar]
- 54.*.Alborghetti MR, Correa MEP, Whangbo J, Shi X, Aricetti JA, da Silva AA, Miranda ECM, Sforca ML, Caldana C, Gerszten RE, et al. : Clinical metabolomics identifies blood serum branched chain amino acids as potential predictive biomarkers for chronic graft vs. host disease. Front. Oncol 2019, 9:141. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this study, authors evaluated 63 metabolites in 18 patients on and observed reduced levels of leucine and isoleucine at day +10 and +100 in patients that go on to develop GVHD, the latter of which was confirmed in validation test set of 49 additional patients.
- 55.**.Liu Y, Huang A, Chen Q, Chen X, Fei Y, Zhao X, Zhang W, Hong Z, Zhu Z, Yang J, et al. : A distinct glycerophospholipid metabolism signature of acute graft versus host disease with predictive value. JCI Insight 2019, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this compelling manuscript, authors combined data from lipid and transcriptomics profiles to reveal an increase in glycerophospholipid (GPL) metabolism. Furthermore, selection of 5 GPL-related metabolites allowed construction of an aGvHD risk score, strongly implicating dysregulated GPL metabolism as an early event in acute GvHD pathogenesis.
- 56.Gibney GT, Weiner LM, Atkins MB: Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016, 17:e542–e551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, Boesteanu AC, Wang Y, O’Connor RS, Hwang W-T, et al. : Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med 2018, 24:563–571. [DOI] [PMC free article] [PubMed] [Google Scholar]