Skip to main content
Immunology logoLink to Immunology
. 2018 Nov 22;156(1):47–55. doi: 10.1111/imm.13016

Aldehyde dehydrogenase in regulatory T‐cell development, immunity and cancer

Christopher G Bazewicz 1,2, Saketh S Dinavahi 2,3, Todd D Schell 4, Gavin P Robertson 2,3,5,6,7,8,9,
PMCID: PMC6283653  PMID: 30387499

Summary

The role of aldehyde dehydrogenase (ALDH) in carcinogenesis and resistance to cancer therapies is well known. Mounting evidence also suggests a potentially important role for ALDH in the induction and function of regulatory T (Treg) cells. Treg cells are important cells of the immune system involved in promoting immune tolerance and preventing aberrant immune responses to beneficial or non‐harmful antigens. However, Treg cells also impair tumor immunity, leading to the progression of various carcinomas. ALDH expression and the subsequent production of retinoic acid by numerous cells, including dendritic cells, macrophages, eosinophils and epithelial cells, seems important in Treg induction and function in multiple organ systems. This is particularly evident in the gastrointestinal tract, pulmonary tract and skin, which are exposed to a myriad of environmental antigens and represent interfaces between the human body and the outside world. Expression of ALDH in Treg cells themselves may also be involved in the proliferation of these cells and resistance to certain cytotoxic therapies. Hence, inhibition of ALDH expression may be useful to treat cancer. Besides the direct effect of ALDH inhibition on carcinogenesis and resistance to cancer therapies, inhibition of ALDH could potentially augment the immune response to tumor antigens by inhibiting Treg induction, function and ability to promote immune tolerance to tumor cells in multiple cancer types.

Keywords: immunotherapy, regulatory T cell, tumor immunology


Abbreviations

ALDH

aldehyde dehydrogenase

CSCs

cancer stem cells

DC

dendritic cell

GM‐CSF

granulocyte–macrophage colony‐stimulating factor

GVHD

graft‐versus‐host disease

IDO

indoleamine 2,3‐dioxygenase

LP

lamina propria

MLN

mesenteric lymph node

MLRs

mixed lymphocyte reactions

PPARγ

peroxisome proliferator‐activated receptor γ

pTreg

peripherally derived regulatory T cells

RA

retinoic acid

SCC

squamous cell carcinoma

TDO

tryptophan dioxygenase

TGF‐β

transforming growth factor‐β

Th

T helper

Treg

regulatory T

Introduction

The aldehyde dehydrogenases (ALDHs) are a superfamily of NADP+‐dependent enzymes that metabolize endogenous and exogenous aldehydes to corresponding carboxylic acids.1 This superfamily of proteins is comprised of 19 isozymes, with constitutive activity of at least one isozyme observed in a majority of mammalian tissues.2 The ALDHs play important roles, among other things, in cellular detoxification, the protection against ultraviolet radiation‐induced damage, and amino acid metabolism.3, 4, 5, 6 Retinoic acid (RA) biosynthesis and signaling (Fig. 1) is another crucial role of the ALDHs, with the conversion of retinal to RA performed by three isozymes in the ALDH1 family, specifically ALDH1A1 (RALDH1), ALDH1A2 (RALDH2) and ALDH1A3 (RALDH3).7, 8, 9 Research also implicates increased ALDH activity within tumors in the pathogenesis of various carcinomas and resistance to cancer therapies.

Figure 1.

Figure 1

Retinoic acid signaling pathway within a cell. Retinol is metabolized to retinal by ADH. ALDH1 (ALDH1A1, ALDH1A2, ALDH1A3) metabolizes retinal to retinoic acid. Retinoic acid binds to RAR (RAR α, RAR β, RAR γ) or RXR in the nucleus. The protein complex then binds to RAREs and modulates gene expression. Abbreviations: ADH, alcohol dehydrogenase; ALDH1, aldehyde dehydrogenase 1; RAR, retinoic acid receptor; RXR, retinoid X receptor; RARE, retinoic acid response element.

Overexpression of various ALDH isozymes in tumor tissue has been associated with a poor prognosis in multiple cancers, including gastric, esophageal, breast, lung, skin, pancreatic, prostate and head and neck squamous cell carcinomas (SCCs).10, 11, 12, 13, 14, 15, 16 Cancer stem cells (CSCs), which are a small subset of multipotent cells within tumors that can drive tumor proliferation, repopulation after various insults (e.g. radiation, chemotherapy, targeted therapy) and metastasis, have elevated ALDH expression.17, 18 CSCs are highly resistant to cancer therapies and can lead to relapse and progression to more aggressive disease.17 Examples include resistance to preoperative chemoradiation in esophageal carcinoma; cytarabine and bortezomib in acute myeloid leukemia; erlotinib, gefitinib and conventional chemotherapy in lung cancer; olaparib in breast cancer; doxorubicin and etoposide in Ewing sarcoma family tumors; cisplatin in mesothelioma and cyclophosphamide in a variety of tumors.16, 19, 20, 21, 22, 23, 24, 25 Hence, inhibition of ALDH has been an active field of research to suppress carcinogenesis and prevent resistance to cancer therapies.

Numerous ALDH inhibitors have been developed in the fight against cancer. These include DEAB, DIMATE, disulfiram, CM026, CM037, NCT‐501, CVT‐10216, ALDH423 and CB29.26, 27, 28, 29, 30, 31, 32, 33 Among these, the broad‐spectrum ALDH inhibitors DEAB, DIMATE and disulfiram either remain in the early stages of development or are limited by their toxicity, while the isoform‐specific ALDH inhibitors CM026, CM037, NCT‐501, CVT‐10216, ALDH423 and CB29 have limited efficacy.26, 34, 35 Developing a broad‐spectrum, non‐toxic ALDH inhibitor would be a major advancement in the cancer field.

In addition to targeting ALDH activity with small molecule inhibitors, direct targeting of ALDH‐positive CSCs through immunological approaches has shown promise in the treatment of cancer.36 For instance, in vitro generation of ADLH1A1‐specific CD8+ T cells with transfer into immunodeficient mice with SCC xenograft led to inhibition of tumor growth and metastases as well as prolonged survival.37 Furthermore, ALDHHigh CSC‐dendritic cell (DC) vaccines have been developed and show efficacy in cancer treatment by inducing targeted cellular and humoral responses to CSCs.38 Vaccination with ALDHHigh CSC‐DCs following localized therapy (e.g. radiation, surgical excision) led to decreased tumor progression, metastasis and prolonged survival in murine models of SCC and melanoma using syngeneic immunocompetent hosts.38 However, these therapeutic approaches are early in development, with more preclinical work needed to establish efficacy and determine toxicity.

Besides the importance of ALDH in carcinogenesis and resistance to cancer therapy, some recent manuscripts suggest that ALDH may play an important role in the immune system. In this review, we present an overview of the current knowledge regarding the role of ALDH in immunity focusing on its effects on regulatory T (Treg) cells. The mounting evidence for the importance of ALDH in Treg induction, function and resistance to cytotoxic therapies is detailed. Finally, the effect of inhibition of ALDH on carcinogenesis is explored to predict possible future research directions that could affect clinical practice.

Role of Treg cells in immunity

Treg cells are immune cells essential for the maintenance of immunological self‐tolerance (e.g. the unresponsiveness of the immune system to self‐antigens).39 They do this through direct cytotoxic effects, the production of anti‐inflammatory cytokines, metabolic disruption and modulation of DC function.40 Deficiency in Treg cell number or function can lead to inflammatory and autoimmune disease. For instance, mutations in the gene encoding Foxp3, a Treg‐specific transcription factor important in Treg cell development, lead to the fatal multi‐organ autoimmune disease immune dysregulation, polyendocrinopathy, enteropathy and X‐linked (IPEX) syndrome.41 Stimulation of Treg cells on the other hand has been shown to help mitigate, and potentially be a treatment option for, numerous autoimmune diseases, such as inflammatory bowel disease.42

Unfortunately, immune tolerance induced by Treg cells against self‐antigens can also impair tumor immunity. Tumor antigens recognized by CD8+ T cells and CD4+ T cells, including Treg cells, are vast and separated into various categories.43 The major categories are: (i) unique antigens, which result from somatic mutations within tumors in ubiquitously expressed genes; (ii) shared antigens, which can be expressed, to various degrees depending on the antigen, in multiple tumor types and in normal tissue; and (iii) viral antigens, which are expressed in virus‐induced malignancies.43 Unlike the anti‐tumor effect of tumor‐infiltrating CD8+ T cells and some CD4+ T‐cell subsets, such as CD4+ T helper type 1 (Th1) cells, the presence of a large number of Treg cells in tumor tissue is generally associated with a pro‐tumor effect, disease progression and subsequently a poorer prognosis.39, 43, 44

Large numbers of tumor‐infiltrating Treg cells have been found in many cancers, including tumors of the ovary, head and neck, pancreas, gastrointestinal tract, liver, lung and breast.45, 46, 47, 48, 49, 50, 51, 52, 53 Increased ratios of tumor‐infiltrating FOXP3+ Treg cells to CD8+ T cells are associated with a poor prognosis, especially for patients with ovarian, gastric and breast carcinomas.50, 52, 53, 54 Similarly, survival for patients with melanoma, and tumors of the breast, kidney and cervix, is significantly reduced when a large number of FOXP3+ Treg cells are present in the tumor.55 Early studies in mice showed that depletion of Treg cells led to the effective eradication of a variety of inoculated syngeneic tumors, partly due to tumor‐specific CD8+ T cells.56 Re‐challenge with the same tumor cells in these mice led to more rapid eradication of the tumor cells, demonstrating the establishment of tumor‐specific immunity.56 Subsequently, numerous therapeutic strategies have been developed to inhibit Treg cells in the hopes of treating various cancers more effectively.

Cellular proteins that have been investigated and targeted in an attempt to deplete and/or functionally impair Treg cells include CD15, CD25, LAG3, OX‐40, PD‐1, CCR4, GITR and CTLA‐4.39 Besides the non‐specificity of these molecules to Treg cells, a major concern with these therapies, particularly anti‐CTLA‐4 and anti‐PD‐1 monoclonal antibodies, is the development of serious autoimmunity, such as enterocolitis, dermatitis or autoimmune thyroid disease, which may prompt discontinuation of the therapy.57 Targeted inhibition of indoleamine 2,3‐dioxygenase (IDO) and tryptophan dioxygenase (TDO), enzymes involved in the rate‐limiting step of tryptophan catabolism, has also shown promise in the treatment of cancer. These enzymes are often overexpressed in neoplastic cells, the tumor microenvironment and draining lymph nodes, leading to immunosuppression, partly through up‐regulation of Treg cells, as well as neovascularization and metastasis.58 Accordingly, inhibition of these enzymes leads to tumor immunity through increased CD8+ T effector cells, natural killer cells and reductions in Treg cells and myeloid‐derived suppressor cells.58 IDO/TDO inhibitors (e.g. epacadostat and navoximod) and the tryptophan mimetic, indoximod, have advanced to phase II/III clinical trials and are generally well tolerated.58 However, they have little activity on their own, but rather enhance ‘immunogenic’ chemotherapy or immune checkpoint drugs.58 Hence, novel approaches are still needed for effective targeting of Treg cells in cancer.

ALDH in Treg induction and function

Several recent manuscripts suggest a potentially important role for ALDH in immunity. This research has focused on the role of ALDH in Treg induction, function and resistance to cytotoxic therapy.59, 60, 61, 62 The gastrointestinal tract,59 pulmonary tract60 and skin61 are the main organ systems that have been identified in which Treg cell activity is enhanced through ALDH (Fig. 2). In short, an immunological stimulus leads to, among other things, up‐regulation of ALDH in multiple cells.59, 60, 61 RA produced from ALDH then enhances Treg induction and function from naive CD4+ T cells.59, 60, 61 These antigen‐specific Treg cells then function to promote immune tolerance (Fig. 2).59, 60, 61

Figure 2.

Figure 2

The currently accepted roles of ALDH in the induction and function of Treg cells in the gastrointestinal tract, pulmonary tract and skin. (a) An immunological stimulus leads to the up‐regulation of ALDH in various cells in the gastrointestinal tract. GM‐CSF and RA secreted by LP macrophages helps induce ALDH in intestinal DCs. RA produced by intestinal DCs, LP eosinophils and macrophages up‐regulates Treg cell number and function, leading to immune tolerance. (b) An immunological stimulus leads to the up‐regulation of ALDH in various cells in the pulmonary tract. GM‐CSF and RA secreted by airway epithelium may help induce ALDH in pulmonary DCs. RA produced by pulmonary DCs, alveolar and resident tissue macrophages up‐regulates Treg cell number, leading to immune tolerance. (c) An immunological stimulus leads to the up‐regulation of ALDH in various cells in the skin. GM‐CSF and RA secreted by fibroblasts, keratinocytes and hair follicles may help induce ALDH in skin DCs. RA produced by skin DCs up‐regulates Treg cell number, leading to immune tolerance. Abbreviations: RA, retinoic acid; LP, lamina propria; GM‐CSF, granulocyte–macrophage colony‐stimulating factor; Eo, eosinophil; DC, dendritic cell; Mo, macrophage; Fo, fibroblast; Treg, regulatory T.

ALDH expression in gastrointestinal immunity

Much evidence for the role of ALDH in Treg induction and function comes from the study of the gastrointestinal tract.59 The intestinal immune system is exposed to a myriad of beneficial and non‐harmful antigens derived from ingested substances and organisms, such as foods and commensal bacteria.59 Oral tolerance is critical to prevent excess immune responses leading to food allergy and intestinal as well as systemic inflammatory disease.63 Antigen‐specific immune suppression through induction of Treg cells plays an important role in this regard.64, 65 In particular, peripherally derived Treg (pTreg) cells appear necessary for oral tolerance, with a majority of intestinal pTreg cells dependent upon food‐derived antigens.66 Lamina propria (LP) and mesenteric lymph node (MLN) DCs, LP macrophages, LP eosinophils and epithelial cells have the ability to induce Treg cell differentiation and promote immune tolerance, partly through increased expression of ALDH.59, 67, 68

LP DCs capture antigens in the intestinal lumen, migrate to MLNs and present the antigens to T cells, which results in the production of Treg cells specific for the antigen.59 The process of Treg cell induction is enhanced by several factors mediated by DCs including stimulation by transforming growth factor‐β (TGF‐β), IDO and RA.69, 70, 71 Both LP and MLN DCs highly express the gene Aldh1a2, which encodes RALDH2.59 RA produced by intestinal DCs also aids in Treg cell function by inducing the gut‐homing receptors CCR9 and integrin α 4 β 7 on T cells, which in turn induces gut‐homing Treg cells able to migrate to the intestine to suppress immune responses to intestinal antigens.72 ALDH produced by intestinal DCs also appears to be important in the intestinal immune tolerance induced by bacteria such as Lactobacillus planatarum, an intestinal bacterium known to have anti‐allergenic and anti‐inflammatory effects.73 In vitro and in vivo studies show induction of Treg cells by L. planatarum through RA‐ and TGF‐β‐mediated intestinal DC stimulation.73

LP macrophages and eosinophils, as well as intestinal epithelium, also aid in the process of Treg induction.68, 74, 75 Evidence suggests thata LP macrophages assist LP DCs by transferring luminal antigens to LP DCs.76 LP macrophages also produce RA through the expression of Aldh1a1 and Aldh1a2 and subsequently RALDH1 and RALDH2, respectively.61, 74 Accordingly, co‐incubation of murine LP macrophages with naive CD4+ T cells induces Treg cell production through interleukin‐10‐ (IL‐10), TGF‐β‐ and RA‐dependent mechanisms.74 Furthermore, LP macrophages produce granulocyte–macrophage colony‐stimulating factor (GM‐CSF), which is important for ALDH induction in intestinal DCs.75 As for eosinophils, co‐incubation of murine LP eosinophils with naive CD4+ T cells induces Treg cell differentiation, which may be important in intestinal immune tolerance, as eosinophils constitute a significant proportion of inflammatory cells within the LP of humans.77 The mechanism by which murine LP eosinophils induce Treg cell differentiation is through expression of ALDH and TGF‐β 1.68 Accordingly, blocking either all‐trans‐RA or TGF‐β 1 inhibits the capability of the eosinophils to induce Treg cells.68 Intestinal epithelial cells contribute to Treg activity, as they are able to produce RA, which is critical for the induction of ALDH in intestinal DCs.75

ALDH expression in pulmonary immunity

Induction of Treg cells by DCs, partly through ALDH, also appears to be involved in immune tolerance in the respiratory tract, which like the gastrointestinal tract is constantly exposed to environmental antigens.60 Lack of pulmonary immune tolerance can lead to unwarranted inflammatory immune responses to inhaled antigens in susceptible individuals, such as asthma.78 Recent studies have shown the importance of pulmonary DCs for inducing Treg cells and subsequently promoting airway tolerance.60, 79, 80, 81 Induction of Treg cells in this setting is largely through TGF‐β production and stimulation of peroxisome proliferator‐activated receptor γ (PPARγ) in DCs.60, 79 PPARγ is a member of the nuclear receptor superfamily with known anti‐inflammatory effects, and is expressed by multiple cell types in the lung, including DCs.82, 83 PPARγ mediates immune tolerance through various mechanisms. The receptor appears to be crucial for expression of Aldh1a2 leading to the production of RA through RALDH2, which along with TGF‐β is necessary for Treg cell induction.60, 84 PPARγ also actively dampens expression of pro‐inflammatory cytokines, such as IL‐6 and IL‐23, which preserves Treg cell function.60 Alveolar and lung‐resident tissue macrophages also express ALDH and are able to induce Treg cells from naive CD4+ T cells via RA and TGF‐β.61, 85, 86 Furthermore, airway epithelium may have the capability to induce ALDH expression in pulmonary DCs as they express Aldh1a1 and potentially could be a source of GM‐CSF.61

ALDH expression in skin immunity

DCs with the capability of Treg induction through ALDH expression and RA production are also present in the skin and draining cutaneous lymph nodes.61 These DCs have increased Aldh1a2 gene expression and the ability to induce skin‐specific Treg cells through RA and TGF‐β.61 Accordingly, inhibition of RA receptors with LE540 led to impaired generation of Treg cells.61 Keratinocytes and fibroblasts may induce ALDH expression in skin DCs as they produce GM‐CSF.61 Furthermore, hair follicles may stimulate ALDH expression in skin DCs by expressing RALDH2 and RALDH3.61 Hence, ALDH expression and the production of RA by skin DCs appear to be important in the cutaneous immune tolerance mediated by Treg cells. Treg cells constitute 5–10% of the T cells in normal skin.40 Deficiency in Treg number or function has been implicated in numerous inflammatory skin diseases, including psoriasis and atopic dermatitis.40

ALDH expression in Treg cells

There is minimal evidence for the role of ALDH action within Treg cells or its effect on Treg activity and function. Most of this evidence comes from work by Kanakry et al. in the study of the immunosuppressive effect of cyclophosphamide following allogeneic blood or bone marrow transplantation (alloBMT).62, 87 Treg cells with elevated ALDH expression may partially explain the success of cyclophosphamide in preventing acute and chronic graft‐versus‐host disease in these patients.62, 87 The utility of cyclophosphamide in preventing GVHD is well established,88 with early evidence suggesting that selective killing of proliferating, alloreactive T cells is the main mechanism by which cyclophosphamide exerts its effect.89 Other studies showed that generation of suppressor T cells following therapy might also contribute to the prevention of GVHD.90 More recent evidence suggests Treg cells play a crucial role in immune tolerance and the prevention of GVHD following post‐transplantation cyclophosphamide.91 Higher pTreg cell numbers are associated with a lower incidence of GVHD in these patients.92 The administration of Treg cells also modulates GVHD and may even be effective in GVHD prophylaxis.93

The mechanism by which Treg cells retain suppressive capability when exposed to cyclophosphamide is hypothesized to be in part through increased expression of ALDH.17, 62, 87 ALDH1 activity is a major pathway through which cyclophosphamide is detoxified in vivo, by oxidation of aldophosphamide to the inactive metabolite, carboxyphosphamide.94 Kanakry et al.62 found that allogeneic stimulation of mixed lymphocyte reactions followed by the administration of mafosfamide, an active cyclophosphamide analogue, led to increased numbers of Treg cells with up‐regulation of ALDH1A1, but not ALDH3A1. Furthermore, treatment of the mixed lymphocyte reactions with DEAB, a competitive ALDH inhibitor, increased sensitivity of Treg cells to mafosfamide.62 Similarly, peripheral blood taken from patients following allogeneic blood or bone marrow transplantation had increased numbers of Treg cells expressing ALDH. The Treg cells were also resistant to treatment with cyclophosphamide.62 Taken together, these results suggest that increased expression of ALDH by Treg cells following post‐transplantation cyclophosphamide may contribute to the immunosuppressive effects of cyclophosphamide in some patients (Fig. 3).

Figure 3.

Figure 3

Role of Treg cell ALDH expression in preventing GVHD in patients receiving post‐transplantation cyclophosphamide. Cyclophosphamide is metabolized by hepatic cytochrome p450 system to 4‐hydroxycyclophosphamide, which equilibrates with aldophosphamide. Both molecules readily diffuse into cells but are not cytotoxic. ALDH1 in Treg cells converts aldophosphamide to the inactive metabolite, carboxyphosphamide. Resistant Treg cells multiply and inhibit GVHD. Abbreviations: ALDH1, aldehyde dehydrogenase 1; Treg, regulatory T; GVHD, graft‐versus‐host disease.

Conclusion

ALDH signaling appears to be increasingly important for Treg cell induction and function through the production of RA by multiple cell types (e.g. DCs, macrophages, eosinophils, epithelial cells) as discussed above. Evidence also suggests that RA is important in Treg cell stability under inflammatory conditions and in inhibiting the differentiation of naive CD4+ T cells into pro‐inflammatory Th17 cells.95, 96 Furthermore, RA may indirectly induce Treg cells through inhibition of memory T cells, which block Treg induction through IL‐4, IL‐21 and interferon‐γ.95, 96 Taken together, ALDH expression and RA‐signaling appear critical for up‐regulating Treg cells and suppressing pathogenic T cells involved in autoimmunity. Also, ALDH expression by Treg cells seems to be critical for resistance of Treg cells to cytotoxic therapies, such as cyclophosphamide.62, 87 However, continued research is needed to further elucidate the contributions of ALDH isozymes and RA signaling, as well as other factors, in the activity and function of Treg cells specifically and immune cells in general.

Treg cells are important immune cells that promote immune tolerance but impair tumor immunity. The T‐cell receptor repertoire of Treg cells is broad, so Treg cells are able to recognize a wide variety of self and tumor antigens.39, 43, 44, 65, 97, 98 Due to the immunosuppressive role of Treg cells, these immune cells have been identified as a target in the treatment of various allergic and autoimmune diseases through Treg stimulation.42 Similarly, treatments aimed at Treg depletion and/or functional impairment are being investigated in various cancers.39

Evidence links ALDH expression by CSCs to the carcinogenesis of various tumors and resistance to cancer therapies. Hence, it is not inconceivable to envision that inhibiting ALDH could have a multipronged effect on cancer by modulating the immune system. Inhibition of ALDH could have a direct anti‐proliferative effect on cancers and increase sensitivity to anti‐cancer drugs or ionizing radiation.19, 26 ALDH inhibition could also increase the ratio of effector T cells to Treg cells within tumor tissue leading to increased tumor immunity.99 For instance, inhibition of RA receptors has been shown to increase the efficacy of anti‐tumor DC vaccines in a murine melanoma model through the suppression of tumor‐infiltrating Treg cells and up‐regulation of tumor‐infiltrating, interferon‐γ‐secreting CD4+ and CD8+ T cells.99 Measurement of the ratios of tumor‐infiltrating lymphocytes within tumor tissue following ALDH inhibition would be needed to confirm a similar mechanism of action.

Research into ALDH inhibitors has been hindered by the fact that broad‐spectrum ALDH inhibitors are generally toxic or remain in early stages of development, and isoform‐specific ALDH inhibitors are generally not effective.26, 34, 35 Hence, the development of a broad‐spectrum, non‐toxic ALDH inhibitor could prove to be a key advancement in this field of cancer research. With the successful development of such a drug, it could potentially be combined with other anti‐cancer drugs and/or radiation therapy to more effectively treat cancer. The sequence in which such an inhibitor is used in combinatorial therapy would need investigation. Anti‐cancer drugs and radiation cause release of tumor antigens and pro‐inflammatory signals, potentially priming or enhancing a Treg cell response.100 Further, Treg cells are more resistant to radiation compared with other T cells, with relative increases seen after radiation.101, 102 Hence, early administration of such an inhibitor in the context of combinatorial therapy could maximize inhibition of the pro‐tumor effect of Treg cells and enhancement of T‐cell‐mediated tumor rejection.

Disclosures

None.

Funding

None.

Author contributions

CGB drafted and edited the manuscript, SSD drafted and edited the manuscript, TDS was involved in project conception and revised the manuscript critically for important intellectual content, and GPR was involved in project conception and revised the manuscript critically for important intellectual content.

References

  • 1. Marchitti SA, Brocker C, Stagos D, Vasiliou V. Non‐P450 aldehyde oxidizing enzymes: the aldehyde dehydrogenase superfamily. Expert Opin Drug Metab Toxicol 2008; 4:697–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Jackson B, Brocker C, Thompson DC, Black W, Vasiliou K, Nebert DW et al Update on the aldehyde dehydrogenase gene (ALDH) superfamily. Hum Genomics 2011; 5:283–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Sladek NE. Human aldehyde dehydrogenases: potential pathological, pharmacological, and toxicological impact. J Biochem Mol Toxicol 2003; 17:7–23. [DOI] [PubMed] [Google Scholar]
  • 4. Seitz HK, Stickel F. Acetaldehyde as an underestimated risk factor for cancer development: role of genetics in ethanol metabolism. Genes Nutr 2010; 5:121–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Brocker C, Lassen N, Estey T, Pappa A, Cantore M, Orlova VV et al Aldehyde dehydrogenase 7A1 (ALDH7A1) is a novel enzyme involved in cellular defense against hyperosmotic stress. J Biol Chem 2010; 285:18452–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Kardos GR, Wastyk HC, Robertson GP. Disruption of proline synthesis in melanoma inhibits protein production mediated by the GCN2 pathway. Mol Cancer Res 2015; 13:1408–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Kedishvili NY. Retinoic acid synthesis and degradation. Subcell Biochem 2016; 81:127–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Roeske TC, Scharff C, Olson CR, Nshdejan A, Mello CV. Long‐distance retinoid signaling in the zebra finch brain. PLoS One 2014; 9:e111722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Brown GT, Cash BG, Blihoghe D, Johansson P, Alnabulsi A, Murray GI. The expression and prognostic significance of retinoic acid metabolising enzymes in colorectal cancer. PLoS One 2014; 9:e90776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Luo Y, Dallaglio K, Chen Y, Robinson WA, Robinson SE, McCarter MD et al ALDH1A isozymes are markers of human melanoma stem cells and potential therapeutic targets. Stem Cells 2012; 30:2100–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Patel M, Lu L, Zander DS, Sreerama L, Coco D, Moreb JS. ALDH1A1 and ALDH3A1 expression in lung cancers: correlation with histologic type and potential precursors. Lung Cancer 2008; 59:340–9. [DOI] [PubMed] [Google Scholar]
  • 12. Marcato P, Dean CA, Giacomantonio CA, Lee PW. Aldehyde dehydrogenase: its role as a cancer stem cell marker comes down to the specific isoform. Cell Cycle 2011; 10:1378–84. [DOI] [PubMed] [Google Scholar]
  • 13. Zheng R, Wang J, Wu Q, Wang Z, Ou Y, Ma L et al Expression of ALDH1 and TGFβ2 in benign and malignant breast tumors and their prognostic implications. Int J Clin Exp Pathol 2014; 7:4173–83. [PMC free article] [PubMed] [Google Scholar]
  • 14. Hoshino Y, Nishida J, Katsuno Y, Koinuma D, Aoki T, Kokudo N et al Smad4 decreases the population of pancreatic cancer‐initiating cells through transcriptional repression of ALDH1A1. Am J Pathol 2015; 185:1457–70. [DOI] [PubMed] [Google Scholar]
  • 15. Matsika A, Srinivasan B, Day C, Mader SA, Kiernan DM, Broomfield A et al Cancer stem cell markers in prostate cancer: an immunohistochemical study of ALDH1, SOX2 and EZH2. Pathology 2015; 47:622–8. [DOI] [PubMed] [Google Scholar]
  • 16. Ajani JA, Wang X, Song S, Suzuki A, Taketa T, Sudo K et al ALDH‐1 expression levels predict response or resistance to preoperative chemoradiation in resectable esophageal cancer patients. Mol Oncol 2014; 8:142–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Muzio G, Maggiora M, Paiuzzi E, Oraldi M, Canuto RA. Aldehyde dehydrogenases and cell proliferation. Free Radic Biol Med 2012; 52:735–46. [DOI] [PubMed] [Google Scholar]
  • 18. Vlashi E, Pajonk F. Cancer stem cells, cancer cell plasticity and radiation therapy. Semin Cancer Biol 2015; 31:28–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Januchowski R, Wojtowicz K, Zabel M. The role of aldehyde dehydrogenase (ALDH) in cancer drug resistance. Biomed Pharmacother 2013; 67:669–80. [DOI] [PubMed] [Google Scholar]
  • 20. Moreb JS, Mohuczy D, Ostmark B, Zucali JR. RNAi‐mediated knockdown of aldehyde dehydrogenase class‐1A1 and class‐3A1 is specific and reveals that each contributes equally to the resistance against 4‐hydroperoxycyclophosphamide. Cancer Chemother Pharmacol 2007; 59:127–36. [DOI] [PubMed] [Google Scholar]
  • 21. Bista R, Lee DW, Pepper OB, Azorsa DO, Arceci RJ, Aleem E. Disulfiram overcomes bortezomib and cytarabine resistance in Down‐syndrome‐associated acute myeloid leukemia cells. J Exp Clin Cancer Res 2017; 36:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Huang CP, Tsai MF, Chang TH, Tang WC, Chen SY, Lai HH et al ALDH‐positive lung cancer stem cells confer resistance to epidermal growth factor receptor tyrosine kinase inhibitors. Cancer Lett 2013; 328:144–51. [DOI] [PubMed] [Google Scholar]
  • 23. Gilabert M, Launay S, Ginestier C, Bertucci F, Audebert S, Pophillat M et al Poly(ADP‐ribose) polymerase 1 (PARP1) overexpression in human breast cancer stem cells and resistance to olaparib. PLoS One 2014; 9:e104302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Cortes‐Dericks L, Froment L, Boesch R, Schmid RA, Karoubi G. Cisplatin‐resistant cells in malignant pleural mesothelioma cell lines show ALDHhighCD44+ phenotype and sphere‐forming capacity. BMC Cancer 2014; 14:304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Awad O, Yustein JT, Shah P, Gul N, Katuri V, O'Neill A et al High ALDH activity identifies chemotherapy‐resistant Ewing's sarcoma stem cells that retain sensitivity to EWS‐FLI1 inhibition. PLoS One 2010; 5:e13943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Koppaka V, Thompson DC, Chen Y, Ellermann M, Nicolaou KC, Juvonen RO et al Aldehyde dehydrogenase inhibitors: a comprehensive review of the pharmacology, mechanism of action, substrate specificity, and clinical application. Pharmacol Rev 2012; 64:520–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Fournet G, Martin G, Quash G. α,β‐acetylenic amino thiolester inhibitors of aldehyde dehydrogenases 1&3: suppressors of apoptogenic aldehyde oxidation and activators of apoptosis. Curr Med Chem 2013; 20:527–33. [DOI] [PubMed] [Google Scholar]
  • 28. Kimble‐Hill AC, Parajuli B, Chen CH, Mochly‐Rosen D, Hurley TD. Development of selective inhibitors for aldehyde dehydrogenases based on substituted indole‐2,3‐diones. J Med Chem 2014; 57:714–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Morgan CA, Hurley TD. Characterization of two distinct structural classes of selective aldehyde dehydrogenase 1A1 inhibitors. J Med Chem 2015; 58:1964–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Yang SM, Yasgar A, Miller B, Lal‐Nag M, Brimacombe K, Hu X et al Discovery of NCT‐501, a potent and selective theophylline‐based inhibitor of aldehyde dehydrogenase 1A1 (ALDH1A1). J Med Chem 2015; 58:5967–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Parajuli B, Georgiadis TM, Fishel ML, Hurley TD. Development of selective inhibitors for human aldehyde dehydrogenase 3A1 (ALDH3A1) for the enhancement of cyclophosphamide cytotoxicity. ChemBioChem 2014; 15:701–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Overstreet DH, Knapp DJ, Breese GR, Diamond I. A selective ALDH‐2 inhibitor reduces anxiety in rats. Pharmacol Biochem Behav 2009; 94:255–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Wang B, Buchman CD, Li L, Hurley TD, Meroueh SO. Enrichment of chemical libraries docked to protein conformational ensembles and application to aldehyde dehydrogenase 2. J Chem Inf Model 2014; 54:2105–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Luo M, Gates KS, Henzl MT, Tanner JJ. Diethylaminobenzaldehyde is a covalent, irreversible inactivator of ALDH7A1. ACS Chem Biol 2015; 10:693–7. [DOI] [PubMed] [Google Scholar]
  • 35. Parajuli B, Fishel ML, Hurley TD. Selective ALDH3A1 inhibition by benzimidazole analogues increase mafosfamide sensitivity in cancer cells. J Med Chem 2014; 57:449–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Pan Q, Li Q, Liu S, Ning N, Zhang X, Xu Y et al Concise Review: Targeting cancer stem cells using immunologic approaches. Stem Cells 2015; 33:2085–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Visus C, Wang Y, Lozano‐Leon A, Ferris RL, Silver S, Szczepanski MJ et al Targeting ALDHbright human carcinoma‐initiating cells with ALDH1A1‐specific CD8(+) T cells. Clin Cancer Res 2011; 17:6174–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Lu L, Tao H, Chang AE, Hu Y, Shu G, Chen Q et al Cancer stem cell vaccine inhibits metastases of primary tumors and induces humoral immune responses against cancer stem cells. Oncoimmunology 2015; 4:e990767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res 2017; 27:109–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Nedoszytko B, Lange M, Sokolowska‐Wojdylo M, Renke J, Trzonkowski P, Sobjanek M et al The role of regulatory T cells and genes involved in their differentiation in pathogenesis of selected inflammatory and neoplastic skin diseases. Part II: The Treg role in skin diseases pathogenesis. Postepy Dermatol Alergol 2017; 34:405–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell 2008; 133:775–87. [DOI] [PubMed] [Google Scholar]
  • 42. Weinhage T, Dabritz J, Brockhausen A, Wirth T, Bruckner M, Belz M et al Granulocyte macrophage colony‐stimulating factor‐activated CD39+/CD73+ murine monocytes modulate intestinal inflammation via induction of regulatory T cells. Cell Mol Gastroenterol Hepatol 2015; 1:433–49.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Protti MP, De Monte L, Di Lullo G. Tumor antigen‐specific CD4+ T cells in cancer immunity: from antigen identification to tumor prognosis and development of therapeutic strategies. Tissue Antigens 2014; 83:237–46. [DOI] [PubMed] [Google Scholar]
  • 44. Fridman WH, Pages F, Sautes‐Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012; 12:298–306. [DOI] [PubMed] [Google Scholar]
  • 45. Schaefer C, Kim GG, Albers A, Hoermann K, Myers EN, Whiteside TL. Characteristics of CD4+CD25+ regulatory T cells in the peripheral circulation of patients with head and neck cancer. Br J Cancer 2005; 92:913–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Liyanage UK, Moore TT, Joo HG, Tanaka Y, Herrmann V, Doherty G et al Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J Immunol 2002; 169:2756–61. [DOI] [PubMed] [Google Scholar]
  • 47. Wolf AM, Wolf D, Steurer M, Gastl G, Gunsilius E, Grubeck‐Loebenstein B. Increase of regulatory T cells in the peripheral blood of cancer patients. Clin Cancer Res 2003; 9:606–12. [PubMed] [Google Scholar]
  • 48. Ormandy LA, Hillemann T, Wedemeyer H, Manns MP, Greten TF, Korangy F. Increased populations of regulatory T cells in peripheral blood of patients with hepatocellular carcinoma. Cancer Res 2005; 65:2457–64. [DOI] [PubMed] [Google Scholar]
  • 49. Ichihara F, Kono K, Takahashi A, Kawaida H, Sugai H, Fujii H. Increased populations of regulatory T cells in peripheral blood and tumor‐infiltrating lymphocytes in patients with gastric and esophageal cancers. Clin Cancer Res 2003; 9:4404–8. [PubMed] [Google Scholar]
  • 50. Sasada T, Kimura M, Yoshida Y, Kanai M, Takabayashi A. CD4+CD25+ regulatory T cells in patients with gastrointestinal malignancies: possible involvement of regulatory T cells in disease progression. Cancer 2003; 98:1089–99. [DOI] [PubMed] [Google Scholar]
  • 51. Hiraoka N, Onozato K, Kosuge T, Hirohashi S. Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clin Cancer Res 2006; 12:5423–34. [DOI] [PubMed] [Google Scholar]
  • 52. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P et al Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004; 10:942–9. [DOI] [PubMed] [Google Scholar]
  • 53. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F et al Intraepithelial CD8+ tumor‐infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A 2005; 102:18538–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL et al Quantification of regulatory T cells enables the identification of high‐risk breast cancer patients and those at risk of late relapse. J Clin Oncol 2006; 24:5373–80. [DOI] [PubMed] [Google Scholar]
  • 55. Shang B, Liu Y, Jiang SJ, Liu Y. Prognostic value of tumor‐infiltrating FoxP3+ regulatory T cells in cancers: a systematic review and meta‐analysis. Sci Rep 2015; 5:15179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. J Immunol 1999; 163:5211–8. [PubMed] [Google Scholar]
  • 57. Larkin J, Chiarion‐Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD et al Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med 2015; 373:23–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Prendergast GC, Malachowski WJ, Mondal A, Scherle P, Muller AJ. Indoleamine 2,3‐dioxygenase and its therapeutic inhibition in cancer. Int Rev Cell Mol Biol 2018; 336:175–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Shiokawa A, Kotaki R, Takano T, Nakajima‐Adachi H, Hachimura S. Mesenteric lymph node CD11b CD103+ PD‐L1High dendritic cells highly induce regulatory T cells. Immunology 2017; 152:52–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Khare A, Chakraborty K, Raundhal M, Ray P, Ray A. Cutting Edge: Dual function of PPARγ in CD11c+ cells ensures immune tolerance in the airways. J Immunol 2015; 195:431–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Guilliams M, Crozat K, Henri S, Tamoutounour S, Grenot P, Devilard E et al Skin‐draining lymph nodes contain dermis‐derived CD103 dendritic cells that constitutively produce retinoic acid and induce Foxp3+ regulatory T cells. Blood 2010; 115:1958–68. [DOI] [PubMed] [Google Scholar]
  • 62. Kanakry CG, Ganguly S, Zahurak M, Bolanos‐Meade J, Thoburn C, Perkins B et al Aldehyde dehydrogenase expression drives human regulatory T cell resistance to posttransplantation cyclophosphamide. Sci Transl Med 2013; 5:211ra157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Steele L, Mayer L, Berin MC. Mucosal immunology of tolerance and allergy in the gastrointestinal tract. Immunol Res 2012; 54:75–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Hori S. Lineage stability and phenotypic plasticity of Foxp3+ regulatory T cells. Immunol Rev 2014; 259:159–72. [DOI] [PubMed] [Google Scholar]
  • 65. Morikawa H, Sakaguchi S. Genetic and epigenetic basis of Treg cell development and function: from a FoxP3‐centered view to an epigenome‐defined view of natural Treg cells. Immunol Rev 2014; 259:192–205. [DOI] [PubMed] [Google Scholar]
  • 66. Kim KS, Hong SW, Han D, Yi J, Jung J, Yang BG et al Dietary antigens limit mucosal immunity by inducing regulatory T cells in the small intestine. Science 2016; 351:858–63. [DOI] [PubMed] [Google Scholar]
  • 67. Scott CL, Aumeunier AM, Mowat AM. Intestinal CD103+ dendritic cells: master regulators of tolerance? Trends Immunol 2011; 32:412–9. [DOI] [PubMed] [Google Scholar]
  • 68. Chen HH, Sun AH, Ojcius DM, Hu WL, Ge YM, Lin X et al Eosinophils from murine lamina propria induce differentiation of naive T cells into regulatory T cells via TGF‐β1 and retinoic acid. PLoS One 2015; 10:e0142881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Coombes JL, Siddiqui KR, Arancibia‐Carcamo CV, Hall J, Sun CM, Belkaid Y et al A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF‐β and retinoic acid‐dependent mechanism. J Exp Med 2007; 204:1757–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M et al Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science 2007; 317:256–60. [DOI] [PubMed] [Google Scholar]
  • 71. Matteoli G, Mazzini E, Iliev ID, Mileti E, Fallarino F, Puccetti P et al Gut CD103+ dendritic cells express indoleamine 2,3‐dioxygenase which influences T regulatory/T effector cell balance and oral tolerance induction. Gut 2010; 59:595–604. [DOI] [PubMed] [Google Scholar]
  • 72. Benson MJ, Pino‐Lagos K, Rosemblatt M, Noelle RJ. All‐trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co‐stimulation. J Exp Med 2007; 204:1765–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Yoshida T, Enomoto M, Nakayama S, Adachi Y, Fujiwara W, Sugiyama H et al Induction of ALDH activity in intestinal dendritic cells by Lactobacillus plantarum NRIC0380. Biosci Biotechnol Biochem 2013; 77:1826–31. [DOI] [PubMed] [Google Scholar]
  • 74. Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17‐producing T cell responses. Nat Immunol 2007; 8:1086–94. [DOI] [PubMed] [Google Scholar]
  • 75. Yokota A, Takeuchi H, Maeda N, Ohoka Y, Kato C, Song SY et al GM‐CSF and IL‐4 synergistically trigger dendritic cells to acquire retinoic acid‐producing capacity. Int Immunol 2009; 21:361–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Mazzini E, Massimiliano L, Penna G, Rescigno M. Oral tolerance can be established via gap junction transfer of fed antigens from CX3CR1+ macrophages to CD103+ dendritic cells. Immunity 2014; 40:248–61. [DOI] [PubMed] [Google Scholar]
  • 77. Rothenberg ME, Mishra A, Brandt EB, Hogan SP. Gastrointestinal eosinophils. Immunol Rev 2001; 179:139–55. [DOI] [PubMed] [Google Scholar]
  • 78. Lambrecht BN, Hammad H. Lung dendritic cells in respiratory viral infection and asthma: from protection to immunopathology. Annu Rev Immunol 2012; 30:243–70. [DOI] [PubMed] [Google Scholar]
  • 79. Khare A, Krishnamoorthy N, Oriss TB, Fei M, Ray P, Ray A. Cutting edge: inhaled antigen upregulates retinaldehyde dehydrogenase in lung CD103+ but not plasmacytoid dendritic cells to induce Foxp3 de novo in CD4+ T cells and promote airway tolerance. J Immunol 2013; 191:25–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Ostroukhova M, Seguin‐Devaux C, Oriss TB, Dixon‐McCarthy B, Yang L, Ameredes BT et al Tolerance induced by inhaled antigen involves CD4+ T cells expressing membrane‐bound TGF‐β and FOXP3. J Clin Invest 2004; 114:28–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Curotto de Lafaille MA, Kutchukhidze N, Shen S, Ding Y, Yee H, Lafaille JJ. Adaptive Foxp3+ regulatory T cell‐dependent and ‐independent control of allergic inflammation. Immunity 2008; 29:114–26. [DOI] [PubMed] [Google Scholar]
  • 82. Wahli W, Michalik L. PPARs at the crossroads of lipid signaling and inflammation. Trends Endocrinol Metab 2012; 23:351–63. [DOI] [PubMed] [Google Scholar]
  • 83. Belvisi MG, Hele DJ, Birrell MA. Peroxisome proliferator‐activated receptor γ agonists as therapy for chronic airway inflammation. Eur J Pharmacol 2006; 533:101–9. [DOI] [PubMed] [Google Scholar]
  • 84. Housley WJ, O'Conor CA, Nichols F, Puddington L, Lingenheld EG, Zhu L et al PPARγ regulates retinoic acid‐mediated DC induction of Tregs. J Leukoc Biol 2009; 86:293–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Coleman MM, Ruane D, Moran B, Dunne PJ, Keane J, Mills KH. Alveolar macrophages contribute to respiratory tolerance by inducing FoxP3 expression in naive T cells. Am J Respir Cell Mol Biol 2013; 48:773–80. [DOI] [PubMed] [Google Scholar]
  • 86. Soroosh P, Doherty TA, Duan W, Mehta AK, Choi H, Adams YF et al Lung‐resident tissue macrophages generate Foxp3+ regulatory T cells and promote airway tolerance. J Exp Med 2013; 210:775–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Ganguly S, Ross DB, Panoskaltsis‐Mortari A, Kanakry CG, Blazar BR, Levy RB et al Donor CD4+ Foxp3+ regulatory T cells are necessary for posttransplantation cyclophosphamide‐mediated protection against GVHD in mice. Blood 2014; 124:2131–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Luznik L, Jones RJ, Fuchs EJ. High‐dose cyclophosphamide for graft‐versus‐host disease prevention. Curr Opin Hematol 2010; 17:493–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Eto M, Mayumi H, Tomita Y, Yoshikai Y, Nishimura Y, Maeda T, et al Specific destruction of host‐reactive mature T cells of donor origin prevents graft‐versus‐host disease in cyclophosphamide‐induced tolerant mice. J Immunol 1991; 146:1402–9. [PubMed] [Google Scholar]
  • 90. Tomita Y, Mayumi H, Eto M, Nomoto K. Importance of suppressor T cells in cyclophosphamide‐induced tolerance to the non‐H‐2‐encoded alloantigens. Is mixed chimerism really required in maintaining a skin allograft tolerance? J Immunol 1990; 144:463–73. [PubMed] [Google Scholar]
  • 91. Hoffmann P, Ermann J, Edinger M, Fathman CG, Strober S. Donor‐type CD4+CD25+ regulatory T cells suppress lethal acute graft‐versus‐host disease after allogeneic bone marrow transplantation. J Exp Med 2002; 196:389–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Magenau JM, Qin X, Tawara I, Rogers CE, Kitko C, Schlough M et al Frequency of CD4+CD25hiFOXP3+ regulatory T cells has diagnostic and prognostic value as a biomarker for acute graft‐versus‐host‐disease. Biol Blood Marrow Transplant 2010; 16:907–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Di Ianni M, Falzetti F, Carotti A, Terenzi A, Castellino F, Bonifacio E et al Tregs prevent GVHD and promote immune reconstitution in HLA‐haploidentical transplantation. Blood 2011; 117:3921–8. [DOI] [PubMed] [Google Scholar]
  • 94. Emadi A, Jones RJ, Brodsky RA. Cyclophosphamide and cancer: golden anniversary. Nat Rev Clin Oncol 2009; 6:638–47. [DOI] [PubMed] [Google Scholar]
  • 95. Raverdeau M, Mills KH. Modulation of T cell and innate immune responses by retinoic Acid. J Immunol 2014; 192:2953–8. [DOI] [PubMed] [Google Scholar]
  • 96. Liu ZM, Wang KP, Ma J, Guo Zheng S. The role of all‐trans retinoic acid in the biology of Foxp3+ regulatory T cells. Cell Mol Immunol 2015; 12:553–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Wooldridge L, Ekeruche‐Makinde J, van den Berg HA, Skowera A, Miles JJ, Tan MP et al A single autoimmune T cell receptor recognizes more than a million different peptides. J Biol Chem 2012; 287:1168–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Birnbaum ME, Mendoza JL, Sethi DK, Dong S, Glanville J, Dobbins J et al Deconstructing the peptide‐MHC specificity of T cell recognition. Cell 2014; 157:1073–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Galvin KC, Dyck L, Marshall NA, Stefanska AM, Walsh KP, Moran B et al Blocking retinoic acid receptor‐α enhances the efficacy of a dendritic cell vaccine against tumours by suppressing the induction of regulatory T cells. Cancer Immunol Immunother 2013; 62:1273–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Demaria S, Golden EB, Formenti SC. Role of local radiation therapy in cancer immunotherapy. JAMA Oncol 2015; 1:1325–32. [DOI] [PubMed] [Google Scholar]
  • 101. Komatsu N, Hori S. Full restoration of peripheral Foxp3+ regulatory T cell pool by radioresistant host cells in scurfy bone marrow chimeras. Proc Natl Acad Sci U S A 2007; 104:8959–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Schaue D, Xie MW, Ratikan JA, McBride WH. Regulatory T cells in radiotherapeutic responses. Front Oncol 2012; 2:90. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Immunology are provided here courtesy of British Society for Immunology

RESOURCES