The regulation of the interaction between the immune system and antigens, which may lead to the induction of immune tolerance, is critical both under physiologic conditions and in different pathological settings. In the past few years, major strides have been made in our understanding of the molecular and cellular bases of this process. Novel pathways have been identified and several novel therapeutic agents are currently under clinical investigation for those diseases in which the normal balance between activation and suppression of the immune response is altered. The tryptophan catabolic enzyme, indoleamine 2,3-dioxygenase (IDO), is one of the key players involved in the inhibition of cell proliferation, including that of activated T cells. Recent works have demonstrated a crucial role for IDO in the induction of immune tolerance during infection, pregnancy, transplantation, autoimmunity, and neoplasias, including hematologic malignancies. In this review, the role of IDO in the induction of immunologic tolerance is addressed with a specific focus on its recently discovered effect on hematologic malignancies.

Indoleamine 2,3-dioxygenase (IDO) is an intracellular heme-containing enzyme that catalyzes the initial rate-limiting step in tryptophan degradation along the kynurenine pathway.1  Tryptophan starvation by IDO consumption inhibits T-cell activation,1,3  whereas products of tryptophan catabolism, such as kynurenine derivatives and O2 free radicals, regulate T-cell proliferation and survival.1,4  IDO is widely expressed in human tissues and cell subsets and is induced during inflammation by IFN-γ and other inflammatory cytokines.5  Recent works have demonstrated a crucial role for IDO in the induction of immune tolerance during infection, pregnancy, transplantation, autoimmunity, and neoplasias, including hematologic malignancies.1,6,7 

In the hematology field, regulatory dendritic cells (DCs) and bone marrow (BM)–derived mesenchymal stem cells (MSCs) expressing IDO have the capacity to suppress T-cell responses to autoantigens and alloantigens.8,9  Moreover, acute myeloid leukemia (AML) cells have been recently demonstrated to express a functionally active form of the IDO protein, which is capable of inhibiting the T-cell response through the expansion of regulatory T cells (Tregs).10 

Although several reviews addressing the enzymatic activity of IDO have recently been published, the aim of the present paper is to summarize the most recent data about IDO expression in hematopoietic cells and its role as a potential therapeutic target in hematologic malignancies.

Tryptophan is an amino acid required by all life forms for protein synthesis and other important metabolic functions, such as the synthesis of the essential cellular factor, nicotinamide adenine dinucleotide (NAD+) and the synthesis of serotonin. Dietary tryptophan is delivered to the liver through the hepatic portal system, and the pool of tryptophan that is not used for protein synthesis can either be degraded in the liver or distributed into the bloodstream where it is captured and used by cells throughout the body. Since the 1970s, several studies have demonstrated that 2 different enzymes initiate the catabolism of tryptophan through a series of metabolic steps known collectively as the kynurenine pathway. Tryptophan 2,3-dioxygenase, or TDO, is an enzyme that is primarily expressed in the liver, is induced by tryptophan and metabolic steroids, and is highly specific for tryptophan. In contrast, IDO is found in many tissues, is induced by inflammatory stimuli, and has less substrate specificity. More recently, a novel gene with homology to IDO has been reported and then subsequently demonstrated to encode an enzyme that catabolizes tryptophan.11,12  This enzyme has been referred to as IDO-2 on the basis of its structural similarity to IDO (Figure 1). However, recent reports have postulated that the 2 forms of IDO may have a differential spectrum of expression and they may have different responses to IDO inhibitors.13 

Figure 1

The kynurenine pathway of tryptophan metabolism.

Figure 1

The kynurenine pathway of tryptophan metabolism.

Close modal

In the last few years, IDO has been extensively investigated since the demonstration of its critical role in regulating immune tolerance to foreign antigens within tissue microenvironments.1  The regulation of the IDO gene, protein, and enzymatic activity is complex and still not completely elucidated. Physiologically, IDO is expressed by a wide variety of cell types in response to several stimuli. Within the immune system, certain subsets of antigen-presenting cells (APCs), including macrophages and DCs, have the capacity to up- and down-regulate IDO expression in response to external stimuli, depending on the microenvironment (Figure 2).14,16  Proinflammatory signals, such as IFN-γ as well as signals from T cells, are known to induce IDO expression in DCs.17,18  In particular, in murine splenic CD8+ DCs and in human monocyte-derived DCs (Mo-DCs), IDO can be induced by the exposure to IFN-γ through the intermediate modulation of a series of relevant genes, including the Tyropb gene via IRF-8 and the signaling adapter DAP12.16,19  Ligation of CD40 on DCs, after inducing the early production of inflammatory mediators via canonical NF-κB signaling, is also known to up-regulate an anti-inflammatory pathway, including IDO expression, via a noncanonical NF-κB signaling mechanism.20,21  Moreover, both in mouse and human DCs, cytotoxic T lymphocyte–associated antigen 4 (CTLA-4) either in a soluble form (CTLA-4-Ig) or bound to the cell membrane of Tregs activates an IDO-dependent tolerogenic program requiring high levels of IFN-γ.18,22,24  In contrast to the hypothesis that IDO is strictly inducible by inflammatory stimuli, recent reports have clearly demonstrated that certain subsets of human myeloid DCs might constitutively express IDO to suppress allogeneic T-cell immune responses.25,26  Taken together, these data contribute to a move toward the identification of a tolerogenic signature in DCs based on the expression of the IDO gene and protein.16  This platform can be used to investigate the molecular basis for IDO expression in cell types other than DCs, including stem cells and tumor cells.

Figure 2

IDO-mediated tryptophan degradation by DCs results in multiple effects, including inhibition of T-cell proliferation, increased T-cell apoptosis, and de novo formation of Tregs. Proinflammatory signals, such as IFN-γ, as well as signals from T cells are known to induce IDO expression in DCs. In particular, CTLA-4 bound to the cell membrane of Tregs activates an IDO-dependent tolerogenic program in DCs. IDO expression by DCs results in inhibition of T-cell effector function, including proliferation and clonal expansion, and reduced survival. Moreover, IDO-expressing DCs may favor the emergence of CD4+CD25+Foxp3+ Tregs by the expansion/conversion from naive CD25Foxp3 T cells.

Figure 2

IDO-mediated tryptophan degradation by DCs results in multiple effects, including inhibition of T-cell proliferation, increased T-cell apoptosis, and de novo formation of Tregs. Proinflammatory signals, such as IFN-γ, as well as signals from T cells are known to induce IDO expression in DCs. In particular, CTLA-4 bound to the cell membrane of Tregs activates an IDO-dependent tolerogenic program in DCs. IDO expression by DCs results in inhibition of T-cell effector function, including proliferation and clonal expansion, and reduced survival. Moreover, IDO-expressing DCs may favor the emergence of CD4+CD25+Foxp3+ Tregs by the expansion/conversion from naive CD25Foxp3 T cells.

Close modal

IDO was originally described to contribute to maternal-fetal tolerance, which is considered an example of foreign antigens to which the immune system should remain tolerant.27  Thus, IDO involvement in such a pivotal aspect of human reproduction indicates that an IDO-dependent metabolic pathway is part of a very basic, ancestral, and evolutionarily conserved mechanism of immune tolerance. Recent experiments in IDO−/− KO mice were not able to demonstrate a role for IDO in central and homeostatic immune tolerance.28  In contrast, several in vivo reports clearly demonstrate that IDO significantly contributes to acquired peripheral tolerance.29,,32 

Two main hypotheses have been proposed to explain the IDO effect in immune regulation.33  First, the tryptophan depletion theory, which emerges from the idea that starving pathogens of tryptophan may result in their elimination, is based on the demonstration that IDO-mediated depletion of tryptophan from the tissue microenvironment or culture medium leads to reduced proliferation and increased apoptosis of T cells.3,34  In this view, Munn et al proposed that the lack of tryptophan is the mechanism responsible for preventing allogeneic fetal rejection.27  More recently, it has been demonstrated that some of the metabolites derived from tryptophan degradation along the kynurenine pathway are immunosuppressive by causing T-cell apoptosis (tryptophan utilization theory).4,35,36  Interestingly, the stress-responsive kinase, general control nonderepressible 2 (GCN2), has been identified as a signaling molecule that enables T cells to respond to stress conditions created by IDO.37,38  Although still speculative, local tryptophan depletion as well as increased kynurenine metabolites may act by up-regulating GCN2, which results in impaired T-cell function.

Naturally arising CD4+CD25+ Foxp3+ Tregs are known to suppress most types of immune responses, including antitumor immunity.39  IDO is expressed and is functionally active in the placenta, which, in turn, is infiltrated by CD4+CD25+ Tregs.40  Moreover, Candida albicans infection increases the number of Tregs due to an IDO induction in host APCs.41  In human cancers, tumor-draining lymph nodes contain IDO-expressing DCs, which enhance Treg function.14,42,43  More recently, Puccetti and collaborators demonstrated that in a nontumoral mouse model, tryptophan catabolism favors the emergence of CD25+Foxp3+ Tregs by their conversion from CD25Foxp3 cells (Fallarino et al38 ). Taken together, these data support the notion that IDO-mediated tryptophan degradation results in multiple effects, including the inhibition of T-cell proliferation, increased T-cell apoptosis, and de novo induction of Tregs, which all lead to an impairment of the cellular immune response. In this context, DCs, which may up-regulate IDO upon environmental stimuli, are believed to play a critical role (Figure 2).

The contribution of IDO to the control of a T cell–mediated immune response is supported by several studies carried out in a variety of pathological conditions, including infections, autoimmunity, transplantation, and cancer.

Infections

Early studies have demonstrated a role for IDO in the inhibition of viruses and intracellular pathogens, such as Toxoplasma gondii and Chlamydia psittaci.44,45  Moreover, the activity of IDO on the induction of a specific inhibitory environment during chronic infection has recently been revisited. In particular, IFN-dependent expression of IDO by different cell subsets, including DCs, is often seen during infections and represents an innate mechanism of pathogen elimination.46 Listeria monocytogenes infection of the mouse placenta, which leads to granulomatous disease, results in the induction of IDO, which, in turn, plays a role in counteracting the bacterial infection while maintaining a barrier to T cells to prevent fetal rejection.47  Similar observations were made with other granulomatous infections caused by Bartonella henselae and Mycobacterium tuberculosis, where IDO-expressing APCs prevent T cell–mediated granuloma destruction and subsequent pathogen dissemination.48  More recently, a critical role for IDO has been found in the generation of immunologic resistance to fungal infections, including candidiasis and aspergillosis. In this setting, DC expression of IDO, tryptophan catabolites, and Tregs all cooperate in regulating the fine balance between inflammation, which is required for protection, especially at the early pathogen encounter phase, and tolerance, which is necessary to down-modulate potentially overwhelming inflammatory responses.41,49 

Autoimmune diseases

In animal models of autoimmunity, such as experimental autoimmune encephalomyelitis (EAE), a large body of evidence clearly supports the role of IDO-mediated tryptophan catabolite generation in skewing a Th1 into a Th2 immune response, resulting in reduced inflammation and tissue damage. Interestingly, an orally active tryptophan metabolite is able to reverse paralysis in mice with EAE.50  In IDO−/− KO mice, IDO promotes Th2-mediated allergy airway inflammation via unique effects on lung DCs.51  In patients with rheumatoid arthritis (RA), another T cell–mediated autoimmune disorder, synovial DCs have been shown to express a functional form of IDO that inhibits T-cell proliferation.52  IDO up-regulation is induced by IFN-γ, which is also responsible for activation of T cells. It is then conceivable that, in this setting, IDO immunosuppressive activity represents a negative feedback effect induced by hyperactivation of T-cell immunity. This observation is in accordance with the hypothesis that one of the biologic functions of IDO may be a counter-regulatory mechanism to suppress excessive immune activation. From this viewpoint, inhibiting IDO activity in autoimmune diseases may have a detrimental effect and may worsen the clinical manifestations, which are induced by a Th1-mediated immune response.

Th1 cells have long been associated with the pathogenesis of many autoimmune diseases. However, the observation of disease in mice deficient in molecules involved in Th1-cell differentiation raised the possibility that other effector T cells were responsible for inducing autoimmunity. Recently, a new CD4+ effector T-cell subset that produces IL-17 (Th17) has emerged. These cells are characterized as preferential producers of interleukin-17A (IL-17A), IL-17F, IL-21, and IL-22.53  Th17 cells and their effector cytokines mediate host defensive mechanisms to various infections, and are involved in the pathogenesis of many autoimmune diseases. In the context of pathogenic inflammation to fungi, IL-17 and IDO-dependent production of kynurenins have been recently proven to play opposing roles.49  Given the emerging pathogenetic role of Th17 cells in autoimmunity, IDO modulation and the in vivo administration of synthetic kynurenins may represent a novel therapeutic means of contrasting Th17 pathway in this setting.

Transplantation

Recently, the immunosuppressive role of IDO has been widely investigated for the induction of graft tolerance. In several transplant models, increased IDO activity in transplanted cells has been demonstrated to have antirejection properties both in vitro and in vivo.54,56  Several authors reported the capacity of immunosuppressive agents, such as CTLA-4Ig and CD40Ig, to favor an acceptance of the transplanted organ by inducing an immune tolerance.55  As recently demonstrated, much of this effect is mediated through the up-regulation of IDO expression in DCs and endothelial cells, which induce a peripheral tolerogenic pathway via the increase of Tregs.22,55  Moreover, in an MHC-mismatched renal allograft model, spontaneous acceptance of the transplanted organ involves several mechanisms leading to Treg expansion via IDO expression in DCs.56 

Allogeneic hematopoietic stem cell transplantation (HSCT) is often complicated by side effects that derive from the profound alteration of the immune system. Graft-versus-host disease (GVHD) is the main cause of morbidity and mortality after HSCT. In recent years, our knowledge of the pathophysiology of GVHD has increased, but the immunologic mechanisms by which GVHD is regulated both at local and systemic levels are still incompletely understood. Infections and T-cell responses directed against alloantigens are known to induce inflammation, which needs a series of negative feedbacks to be efficiently counterbalanced. Among these, the induction of IDO, which mediates anti-inflammatory activities and T-cell inhibition via tryptophan catabolism, have been postulated.57 

Recently, IDO has been demonstrated to act as a critical regulator of GVHD in a mouse model of HSCT.58  By using an IDO−/− KO mouse model, it has been demonstrated that the absence of IDO results in increased colon GVHD and reduced survival of transplanted tissue in IDO−/− mice compared with wild-type mice. Interestingly, in IDO−/− mice, the major effect is isolated to the site of GVHD, mainly in the gut, with increased infiltration of proliferating CD4+ and CD8+ T cells, whereas there was little to no systemic effect found (ie, activation of T cells in lymphoid organs and/or induction of Tregs). The critical role for IDO expression in orchestrating T-cell function during GVHD at the site of inflammation represents a major advance in our knowledge on the environmental effects influencing GVHD tissue injury and offers a rationale for a novel approach to GVHD management. Interestingly, MSCs, which are currently under clinical investigation for the induction of immunologic tolerance within allogeneic stem cell transplantation,59  have been shown to express a functionally active form of IDO after exposure to IFN-γ.9  This observation provides a rationale for exploiting IDO-mediated tolerance as part of a cell therapy approach to GVHD.

Cancer

During tumor development, cancer cells acquire certain cell-intrinsic characteristics, such as immortalization, growth signal self-sufficiency, and apoptosis resistance, as well as some properties that are defined through the interaction with the host environment (cell extrinsic). Among the latter, the capacity of tumor cells to interact with the immune system is typical and may represent a crucial step in the process of malignant transformation. Indeed, several reports support the hypothesis that the immune system–tumor cell interaction plays a critical and dual role in tumor development both by eliminating tumor cells and by facilitating tumor escape from immune control. Recent evidence has demonstrated that the immune system can readily destroy transformed cells but can also promote the selection of neoplastic clones with reduced immunogenicity, thus providing developing tumors with a mechanism to escape immunologic detection and elimination.60 

Recently, a wide variety of human solid tumors have been demonstrated to express an active IDO protein, and transfection of IDO into tumor cells prevents their rejection by preimmunized hosts.6,43  The antitumor effect of IDO blockade is completely dependent on the presence of a fully competent immune system, thus suggesting that IDO acts by deregulating the host immune response and plays a pivotal role in the inhibition of tumor-specific immunity. The mechanism(s) by which IDO expression leads to the induction of immune tolerance to tumors has been widely investigated in recent years. These studies suggest that IDO protein is critically involved in this process both at the tumor site, where the depletion of tryptophan and the increase of immunosuppressive metabolites results in reduced effector function, clonal expansion, and survival of antigen-specific T cells, and at the level of the tumor-draining lymph node, where a population of IDO-expressing plasmacytoid DCs have a tolerogenic effect on T cells during their encounter with antigen-loaded APCs (Figure 3). Interestingly, IDO-expressing DCs have been observed in sentinel lymph nodes of patients with melanoma and are associated with poor clinical outcome.14 

Figure 3

IDO protein is involved in the induction of tumor tolerance. IDO is up-regulated in both tumor cells (right) and in a subset of regulatory/plasmacytoid DCs within tumor-draining lymph nodes (left). At the tumor site, tryptophan degradation impairs the effector function of antigen-specific T cells and reduces the immune-mediated control of tumor growth. In tumor-draining lymph nodes, regulatory DCs create a tolerogenic microenvironment, where the presentation of relevant tumor antigens to developing T cells may be defective, which leads to reduced numbers of antigen-specific T cells and increased numbers of Tregs.

Figure 3

IDO protein is involved in the induction of tumor tolerance. IDO is up-regulated in both tumor cells (right) and in a subset of regulatory/plasmacytoid DCs within tumor-draining lymph nodes (left). At the tumor site, tryptophan degradation impairs the effector function of antigen-specific T cells and reduces the immune-mediated control of tumor growth. In tumor-draining lymph nodes, regulatory DCs create a tolerogenic microenvironment, where the presentation of relevant tumor antigens to developing T cells may be defective, which leads to reduced numbers of antigen-specific T cells and increased numbers of Tregs.

Close modal

In humans, IDO overexpression has been correlated with poor prognosis in patients with ovarian carcinoma, endometrial carcinoma, and colon carcinoma.61,63  In particular, for colorectal cancer, IDO expression by tumor cells is associated with a reduction in tumor-infiltrating lymphocytes.63  These data are in agreement with the hypothesis that increased IDO activity within a tumor site creates an immunosuppressive microenvironment that inhibits effector antigen-specific T cells. The genetic basis of IDO gene regulation in tumor cells as well as its interplay with other aspects of malignant conversion, such as tumor cell proliferation and apoptosis, is not completely elucidated. One recent report demonstrated that IDO is under the genetic control of BIN1, which acts as a cancer suppressive gene and is down-regulated in many human malignancies.64  In particular, deletion of the BIN1 gene from cells results in the overexpression of IDO gene by IFN-γ. These experimental data may suggest that, at least in some cases, IDO is constitutively expressed by tumor cells as part of the genetic changes involved in malignant transformation and provide a platform to investigate the molecular link between tumor cell transformation and the interaction of tumor cells with the host immune system. Alternatively, since IDO is known to be induced in different cell subsets upon stimulation with IFN-γ, its expression by tumor cells may be a response to inflammatory cytokines, which are produced in response to the initial host immune activation against the tumor.

The leukemogenic process has been widely investigated over the last 3 decades and major advances have been made in our understanding of the molecular and cellular bases of leukemias.65  Moreover, several reports support the notion that the leukemic clone is influenced by the host immune system, which can counteract the development and growth of leukemic cells. Clinical and biologic results from allogeneic HSCT represent the major evidence that leukemia cells are sensitive to the antitumor activity of the immune system. Since the first observation that allogeneic BM transplantation offered a clinical advantage over autologous transplantation, much more attention has been given to the role of adoptive immunotherapy over a conditioning regimen as a means to eradicate tumor cells. In particular, donor lymphocyte infusions (DLIs) in patients with chronic myeloid leukemia (CML) who relapse after HSCT are able to reinduce hematologic and molecular complete remission.66  Moreover, tumor-specific T lymphocytes have been demonstrated to participate in the elimination of CML cells in patients undergoing HSCT or IFN-α–based treatment.67  More recently, patients with AML have been successfully treated with a leukemia-specific vaccine formulation, which resulted in the eradication of minimal residual disease.68  These results, along with the increasing use of so-called mini-transplants, are the proof of principle for the crucial activity of antitumor immunity in controlling leukemia growth.

Along with the demonstration of the capacity of the immune system to recognize and destroy leukemia cells, recent reports have also provided evidence that leukemia cells exert several mechanisms to escape tumor-specific immune responses. AML cells prevent T-cell activation and proliferation both through a cell-to-cell contact mechanism and by producing soluble factors inhibiting the Th1 immune response.69,70  Transgene expression of costimulatory molecules into AML cells is not sufficient to restore T-cell function, thus suggesting that the leukemic microenvironment is very important in inducing the T-cell deficit.70  Moreover, CML cells are known to shape an antileukemia immune response by depleting high-avidity T-cell clones.71  We demonstrated that AML cells, but not their normal counterparts (ie, CD34+ hematopoietic stem/progenitor cells [HSCs]), express an active IDO protein that converts tryptophan into kynurenine and inhibits allogeneic T-cell proliferation.8  The differential expression of IDO among normal CD34+ HSCs and leukemic cells supports the hypothesis that, in a subset of AML patients, the constitutive expression of IDO protein is associated with the leukemic transformation and may play a role in leukemia development. IDO gene expression by leukemic blasts has been recently analyzed using a quantitative reverse-transcription–polymerase chain reaction (RT-PCR) method. Fifty-four newly diagnosed AML patients have been studied. According to the IDO transcript level, AML patients could be subdivided into 2 groups (Figure 4; A.C., unpublished data, February 2009). In particular, a group of patients expressed IDO at the same level as normal BM cells (IDO negative, n = 40), whereas 14 patients (26%) showed a significantly increased IDO mRNA expression at a level comparable with that of the positive control (placenta cells). We and others6  have also shown that there is a direct correlation between the level of mRNA and the presence of detectable amounts of protein, which is measurable by enzymatic activity. Indeed, only leukemic samples with a high expression of IDO mRNA have detectable amounts of protein, which was measured by Western blot analysis and immunohistochemistry, and consequently, show significant production of tryptophan metabolites along the kynurenine pathway.7  The clinical relevance of IDO expression by AML cells is under active clinical investigation by several groups. As mentioned above, IDO expression by solid tumors confers a poor clinical outcome. Very recently, IDO overexpression in AML blasts has been confirmed by other groups.72,,75  IDO activity of AML cells can be measured from patients' sera by high-performance liquid chromatography (HPLC) and is inducible by IFN-γ.72  Moreover, correlation of IDO expression with well-known prognostic factors and survival identified high IDO expression as a strong negative independent predicting variable for overall survival and relapse-free survival.75 

Figure 4

IDO expression by AML cells. IDO gene expression by AML blasts has been analyzed using a quantitative RT-PCR method in 54 newly diagnosed patients. IDO mRNA level was compared with that of Abelson (ABL) control gene. According to the IDO transcript level, AML patients could be subdivided into 2 groups. In particular, a group of patients (n = 40) expressed IDO at the same level as normal BM-derived CD34+ cells (IDO). The remaining samples (n = 14) showed a significantly increased IDO mRNA expression that was at a level comparable with that of the positive control placenta cells (IDO+).

Figure 4

IDO expression by AML cells. IDO gene expression by AML blasts has been analyzed using a quantitative RT-PCR method in 54 newly diagnosed patients. IDO mRNA level was compared with that of Abelson (ABL) control gene. According to the IDO transcript level, AML patients could be subdivided into 2 groups. In particular, a group of patients (n = 40) expressed IDO at the same level as normal BM-derived CD34+ cells (IDO). The remaining samples (n = 14) showed a significantly increased IDO mRNA expression that was at a level comparable with that of the positive control placenta cells (IDO+).

Close modal

A significantly increased number of Tregs have been observed in several tumors, such as lung, breast, pancreatic, ovarian carcinomas, and melanoma, and Tregs have been found to suppress the tumor-specific immune response.76,78  In hematologic malignancies, an increase in circulating CD4+CD25+ Tregs has been demonstrated in Hodgkin lymphoma, non-Hodgkin lymphoma (NHL), chronic lymphocytic leukemia, AML, multiple myeloma, and high-risk myelodysplastic syndromes.79,,,,84  One recent comprehensive survey of Treg frequency in NHL has been published.85  The authors found that all subtypes of lymphomas had increased Tregs and their number was associated with disease stage and other biomarkers correlated with tumor burden (ie, lactate dehydrogenase). Interestingly, there were abnormally high numbers of Tregs in both the peripheral blood and locally involved tissues, thus suggesting that the microenvironment induced by lymphoma cells may be responsible for Treg induction.

The origin of the increased number of Tregs in cancer patients needs further elucidation. Murine tumors of different histologic types are capable of expanding Treg numbers by converting CD4+CD25 into CD25+ T cells.86  It was postulated that tumor-derived soluble factors, such as transforming growth factor-β1,87  may play a critical role in inducing the Treg conversion. Other possibilities include the recruitment and/or expansion of the circulating Treg population within tumoral tissues. We recently showed that AML cells induce in vitro CD4+CD25+Foxp3+ Tregs from the Foxp3CD25 T-cell fraction via expression of IDO protein and modulation of tryptophan catabolism.10  Increased levels of circulating Tregs have been associated with a high expression of IDO protein by AML cells. The same results were obtained using a mouse model of leukemia/lymphoma, where the transgenic overexpression of IDO in tumor cells induced an increase in the Treg frequency in peripheral blood and within lymphatic tissues, such as the spleen and lymph nodes. These data point to a direct role for IDO production in the expansion of converting Tregs by AML cells and are in agreement with the results by Puccetti and collaborators demonstrating that, in a nontumoral mouse model, tryptophan catabolism favors the emergence of CD25+Foxp3+ Tregs by conversion from CD25Foxp3 cells (Fallarino et al38 ). It remains to be elucidated how the modulation of tryptophan catabolism by IDO-expressing AML cells may be implicated in the conversion of CD4+CD25 cells into CD4+CD25+ cells. A tumor microenvironment containing reduced concentrations of tryptophan and high concentrations of kynurenine may play a role in the tumor-mediated induction of Tregs. Interestingly, the accumulation of small molecules, such as l-arginine, nitric oxide, and polyamines, within the tumor microenvironment has recently been demonstrated to affect tumor-infiltrating cell populations, including Tregs.88  More recently, ATP metabolites, which may be generated by Tregs within tumor microenvironment, have been demonstrated to suppress DC maturation and to induce IDO expression through triggering adenosine receptors.89 

Several preclinical studies have demonstrated that IDO inhibition may result in a therapeutic effect, especially in murine models of cancer. IDO inhibitors include small molecules, such as 1-methyl-tryptophan (1-MT), which abrogates IDO enzymatic activity by competing with the normal tryptophan substrate. Initially, 1-MT was used in vivo to test the effect of IDO in tumor cells, which had been genetically modified to overexpress IDO protein.6  These studies demonstrated that 1-MT can slow down the development and growth of IDO-expressing tumor cells, but it cannot induce their regression. Similarly, in the spontaneous mammary tumors that develop in the MMTV-neu/HER2 transgenic mouse model of breast cancer, 1-MT was effective at reducing tumor growth, but did not affect tumor-free survival.64  However, when combined with different types of cytotoxic chemotherapeutic agents, 1-MT induced regression of MMTV-neu/HER2 tumors. This effect was mediated by a T-cell pathway since it was abolished by the depletion of T cells before the treatment. Collectively, these data indicate that the combination of an immunomodulatory agent inhibiting IDO activity and chemotherapeutics may be more effective than the single agents used alone. Indeed, it is well known that cancer chemotherapy may favor antigen presentation by causing the release of tumor antigens from dying tumor cells.90  Moreover, some cytotoxic agents may act by inactivating tumor-protective Tregs, thus increasing the efficacy of immunotherapeutic interventions.91  However, in most studies, chemotherapy is not sufficient to elicit a protective antitumor immune response against cancer. It would be interesting to determine whether inhibiting IDO after chemotherapy would result in better outcomes since it persistently breaks the tolerance to tumors, thus eliciting an effective immune response.

1-MT exists as both d and l isomers. Although d-1-MT has recently entered a phase 1 clinical trial as immune adjuvant for cancer immunotherapy (http://www.clinicaltrial.gov), it is still a matter of debate which isomer would be more powerful as an immune-modulating agent. Based on its chemical structure, it would be expected that only l-1-MT may exert a significant effect in inhibiting IDO enzymatic activity. The superiority of l-1-MT was clearly demonstrated when both molecules were tested to inhibit IDO-expressing cell lines.92  However, when the 2 compounds were compared in mixed lymphocyte reactions to suppress in vitro IDO-mediated tolerogenic effect on T cells, d-1-MT proved superior to l-1-MT.3  In addition, reports using in vivo murine models of cancer demonstrated that d-1-MT inhibited IDO activity better. Interestingly, recent data show that IDO2 is preferentially inhibited by d-1-MT, in contrast to IDO, which is inhibited by l-1-MT.13  Recent reports have demonstrated a differential expression of IDO and IDO2 by different cell subsets. It is then possible that the contrasting results obtained with 1-MT isoforms under different experimental conditions may be a result of the differential expression and susceptibility to inhibition of IDO targets. Other IDO-inhibitor molecules are currently under investigation.93  In particular, the nontoxic inhibitor, methyl-thiohydantoin tryptophan, and the natural product, brassinin, both display great potency in inhibiting IDO activity in a similar way as 1-MT.93,94  Given the promising preclinical results in inhibiting the tolerance induction to tumors, it is of major interest for clinicians to clearly demonstrate which IDO-inhibitor should be used for clinical purposes.

As discussed in this review, IDO has recently emerged as a key regulator of the immune response. IDO activity has been widely investigated and is currently a well-established inducer of the immune tolerance to tumors. Taken together, these data offer a rationale for the clinical investigation of the capacity of IDO inhibitors to increase the efficacy of anticancer immunotherapy, in addition to conventional chemotherapeutic agents. In particular, active vaccination may significantly benefit from a multipronged approach, which includes the destruction of tumor cells by chemotherapy with a subsequent release of tumor antigens and prevention of the IDO-mediated immunologic tolerance. Ongoing and future clinical studies should both address whether IDO suppression significantly impacts the efficacy of immunotherapeutic interventions against cancer and provide evidence to determine whether IDO expression represents a fundamental pathway in tumor development, including hematologic malignancies. Besides tumors, the demonstration of a critical effect for IDO in reducing Th1-mediated tissue damage has significant implications for a variety of clinical settings, including GVHD. GVHD is the main cause of morbidity and mortality after HSCT and novel approaches of GVHD treatment are necessary. The modulation of IDO in GVHD target organs may represent an interesting strategy to limit GVHD by acting at sites where host and donor cells interact, whereas other interventions, such as Treg infusional therapy, may be used to induce systemic T-cell tolerance within lymphoid organs. Future clinical applications of these recent biologic advances are warranted since the interest in developing therapeutic targets against the IDO pathway is expected to be continuously growing.

The authors thank Dr Mario Colombo for his critical review of the paper. The authors apologize to colleagues whose work could not be cited due to space limitations.

This work was supported by Consiglio Nazionale delle Ricerche (CNR, Rome, Italy; N. CU03.00334), Italian Ministry of Health (Progetto Strategico Oncologia, 2006), Regione Emilia-Romagna (Progetti di Ricerca Università-Regione Emilia Romagna, Progetto Medicina Rigenerativa, 2007), Italian Association Against Leukemia, Section of Bologna (BolognAIL), and Ministero dell'Istruzione, dell'Università e della Ricerca (MIUR; PRIN 2006).

Contribution: A.C. designed the research, analyzed the data, and wrote the paper; S.T. and V.S. performed research and analyzed the data; M.B. critically reviewed the paper; and R.M.L. designed the research and critically reviewed the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Antonio Curti, Department of Hematology and Oncological Sciences L & A Seràgnoli, Via Massarenti, 9, 40138, Bologna, Italy; e-mail: antonio.curti2@unibo.it.

1
Mellor
AL
Munn
DH
IDO expression by dendritic cells: tolerance and tryptophan catabolism.
Nat Rev Immunol
2004
4
762
774
2
Mellor
AL
Munn
DH
Tryptophan catabolism and T-cell tolerance: immunosuppression by starvation?
Immunol Today
1999
20
469
473
3
Mellor
AL
Keskin
DB
Johnson
T
Chandler
P
Munn
DH
Cells expressing indoleamine 2,3-dioxygenase inhibit T cell responses.
J Immunol
2002
168
3771
3776
4
Frumento
G
Rotondo
R
Tonetti
M
Damonte
G
Benatti
U
Ferrara
GB
Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase.
J Exp Med
2002
196
459
468
5
Däubener
W
MacKenzie
CR
IFN-gamma activated indoleamine 2,3-dioxygenase activity in human cells is an antiparasitic and an antibacterial effector mechanism.
Adv Exp Med Biol
1999
467
517
524
6
Uyttenhove
C
Pilotte
L
Théate
I
et al
Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase.
Nat Med
2003
9
1269
1274
7
Curti
A
Aluigi
M
Pandolfi
S
et al
Acute myeloid leukemia cells constitutively express the immunoregulatory enzyme indoleamine 2,3-dioxygenase.
Leukemia
2007
21
353
355
8
Munn
DH
Sharma
MD
Lee
JR
et al
Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase.
Science
2002
297
1867
1870
9
Meisel
R
Zibert
A
Laryea
M
Gobel
U
Daubener
W
Dilloo
D
Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation.
Blood
2004
103
4619
4621
10
Curti
A
Pandolfi
S
Valzasina
B
et al
Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25- into CD25+ T regulatory cells.
Blood
2007
109
2871
2877
11
Ball
HJ
Sanchez-Perez
A
Weiser
S
et al
Characterization of an indoleamine 2,3-dioxygenase-like protein found in humans and mice.
Gene
2007
396
203
213
12
Metz
R
Duhadaway
JB
Kamasani
U
Laury-Kleintop
L
Muller
AJ
Prendergast
GC
Novel tryptophan catabolic enzyme IDO2 is the preferred biochemical target of the antitumor indoleamine 2,3-dioxygenase inhibitory compound D-1-methyl-tryptophan.
Cancer Res
2007
67
7082
7087
13
Lob
S
Konigsrainer
A
Schafer
R
Rammensee
HG
Opelz
G
Terness
P
Levo- but not dextro-1-methyl tryptophan abrogates the IDO activity of human dendritic cells.
Blood
2008
111
2152
2154
14
Munn
DH
Sharma
MD
Hou
D
et al
Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes.
J Clin Invest
2004
114
280
290
15
Grohmann
U
Fallarino
F
Bianchi
R
et al
IL-6 inhibits the tolerogenic function of CD8 alpha(+) dendritic cells expressing indoleamine 2,3-dioxygenase.
J Immunol
2001
167
708
714
16
Orabona
C
Puccetti
P
Vacca
C
et al
Toward the identification of a tolerogenic signature in IDO-competent dendritic cells.
Blood
2006
107
2846
2854
17
Taylor
MW
Feng
GS
Relationship between interferon-gamma, indoleamine 2,3-dioxygenase, and tryptophan catabolism.
FASEB J
1991
5
2516
2522
18
Grohmann
U
Bianchi
R
Orabona
C
et al
Functional plasticity of dendritic cell subsets as mediated by CD40 versus B7 activation.
J Immunol
2003
171
2581
2587
19
Romani
L
Bistoni
F
Perruccio
K
et al
Thymosin alpha1 activates dendritic cell tryptophan catabolism and establishes a regulatory environment for balance of inflammation and tolerance.
Blood
2006
108
2265
2274
20
Tas
SW
Vervoordeldonk
MJ
Hajji
N
et al
Noncanonical NF-kappaB signaling in dendritic cells is required for indoleamine 2,3-dioxygenase (IDO) induction and immune regulation.
Blood
2007
110
1540
1549
21
Puccetti
P
Grohmann
U
IDO and regulatory T cells: a role for reverse signalling and non-canonical NF-kappaB activation.
Nat Rev Immunol
2007
7
817
823
22
Grohmann
U
Orabona
C
Fallarino
F
et al
CTLA-4-Ig regulates tryptophan catabolism in vivo.
Nat Immunol
2002
3
1097
1101
23
Fallarino
F
Grohmann
U
Hwang
KW
et al
Modulation of tryptophan catabolism by regulatory T cells.
Nat Immunol
2003
4
1206
1212
24
Munn
DH
Sharma
MD
Mellor
AL
Ligation of B7-1/B7-2 by human CD4+ T cells triggers indoleamine 2,3-dioxygenase activity in dendritic cells.
J Immunol
2004
172
4100
4110
25
Terness
P
Chuang
JJ
Bauer
T
Jiga
L
Opelz
G
Regulation of human auto- and alloreactive T cells by indoleamine 2, 3-dioxygenase (IDO)-producing dendritic cells: too much ado about IDO?
Blood
2005
105
2480
2486
26
Munn
DH
Mellor
AL
Rossi
M
Young
JW
Dendritic cells have the option to express IDO-mediated suppression or not [letter].
Blood
2005
105
2618
27
Munn
DH
Zhou
M
Attwood
JT
et al
Prevention of allogeneic fetal rejection by tryptophan catabolism.
Science
1998
281
1191
1193
28
Mellor
AL
Baban
B
Chandler
P
et al
Cutting edge: induced indoleamine 2,3 dioxygenase expression in dendritic cell subsets suppresses T cell clonal expansion.
J Immunol
2003
17
1652
1655
29
Gurtner
GJ
Newberry
RD
Schloemann
SR
McDonald
KG
Stenson
WF
Inhibition of indoleamine 2,3-dioxygenase augments trinitrobenzene sulfonic acid colitis in mice.
Gastroenterology
2003
125
1762
1773
30
Kwidzinski
E
Bunse
J
Aktas
O
et al
Indolamine 2,3-dioxygenase is expressed in the CNS and down-regulates autoimmune inflammation.
FASEB J
2005
19
1347
1349
31
Hayashi
T
Beck
L
Rossetto
C
et al
Inhibition of experimental asthma by indoleamine 2,3-dioxygenase.
J Clin Invest
2004
114
270
279
32
Beutelspacher
SC
Tan
PH
McClure
MO
Larkin
DF
Lechler
RI
George
AJ
Expression of indoleamine 2,3-dioxygenase (IDO) by endothelial cells: implications for the control of alloresponses.
Am J Transplant
2006
6
1320
1330
33
Moffett
JR
Namboodiri
MA
Tryptophan and the immune response.
Immunol Cell Biol
2003
81
247
265
34
Munn
DH
Shafizadeh
E
Attwood
JT
Bondarev
I
Pashine
A
Mellor
AL
Inhibition of T cell proliferation by macrophage tryptophan catabolism.
J Exp Med
1999
189
1363
1372
35
Fallarino
F
Grohmann
U
Vacca
C
et al
T cell apoptosis by tryptophan catabolism.
Cell Death Differ
2002
9
1069
1077
36
Bauer
TM
Jiga
LP
Chuang
JJ
Randazzo
M
Opelz
G
Terness
P
Studying the immunosuppressive role of indoleamine 2,3-dioxygenase: tryptophan metabolites suppress rat allogeneic T-cell responses in vitro and in vivo.
Transpl Int
2005
18
95
100
37
Munn
DH
Sharma
MD
Baban
B
et al
GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase.
Immunity
2005
22
633
642
38
Fallarino
F
Grohmann
U
You
S
et al
The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T cell receptor zeta-chain and induce a regulatory phenotype in naive T cells.
J Immunol
2006
176
6752
6761
39
O'Garra
A
Vieira
P
Regulatory T cells and the mechanisms of immune system control.
Nat Med
2004
10
801
805
40
Kudo
Y
Boyd
CA
Spyropoulou
I
et al
Indoleamine 2,3-dioxygenase: distribution and function in the developing human placenta.
J Reprod Immunol
2004
61
87
98
41
Bozza
S
Fallarino
F
Pitzurra
L
et al
A crucial role for tryptophan catabolism at the host/Candida albicans interface.
J Immunol
2005
174
2910
2918
42
Sharma
MD
Baban
B
Chandler
P
et al
Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase.
J Clin Invest
2007
117
2570
2582
43
Munn
DH
Mellor
AL
Indoleamine 2,3-dioxygenase and tumor-induced tolerance.
J Clin Invest
2007
117
1147
1154
44
Yoshida
R
Urade
Y
Tokuda
M
Hayaishi
O
Induction of indoleamine 2,3-dioxygenase in mouse lung during virus infection.
Proc Natl Acad Sci U S A
1979
76
4084
4086
45
Yoshida
R
Hayaishi
O
Induction of pulmonary indoleamine 2,3-dioxygenase by intraperitoneal injection of bacterial lipopolysaccharide.
Proc Natl Acad Sci U S A
1978
75
3998
4000
46
Däubener
W
Spors
B
Hucke
C
et al
Restriction of Toxoplasma gondii growth in human brain microvascular endothelial cells by activation of indoleamine 2,3-dioxygenase.
Infect Immun
2001
69
6527
6531
47
Mackler
AM
Barber
EM
Takikawa
O
Pollard
JW
Indoleamine 2,3-dioxygenase is regulated by IFN-gamma in the mouse placenta during Listeria monocytogenes infection.
J Immunol
2003
170
823
830
48
Kaufmann
SH
Immunity to intracellular bacteria.
Annu Rev Immunol
1993
11
129
163
49
Romani
L
Zelante
T
De Luca
A
Fallarino
F
Puccetti
P
IL-17 and therapeutic kynurenines in pathogenic inflammation to fungi.
J Immunol
2008
180
5157
5162
50
Platten
M
Ho
PP
Youssef
S
et al
Treatment of autoimmune neuroinflammation with a synthetic tryptophan metabolite.
Science
2005
31
850
855
51
Xu
H
Oriss
TB
Fei
M
et al
Indoleamine 2,3-dioxygenase in lung dendritic cells promotes Th2 responses and allergic inflammation.
Proc Natl Acad Sci U S A
2008
105
6690
6695
52
Zhu
L
Ji
F
Wang
Y
et al
Synovial autoreactive T cells in rheumatoid arthritis resist IDO-mediated inhibition.
J Immunol
2006
177
8226
8233
53
Dong
C
TH17 cells in development: an updated view of their molecular identity and genetic programming.
Nat Rev Immunol
2008
8
337
348
54
Laurence
JM
Wang
C
Park
ET
et al
Blocking indoleamine dioxygenase activity early after rat liver transplantation prevents long-term survival but does not cause acute rejection.
Transplantation
2008
85
1357
1361
55
Guillonneau
C
Hill
M
Hubert
FX
et al
CD40Ig treatment results in allograft acceptance mediated by CD8CD45RC T cells, IFN-gamma, and indoleamine 2,3-dioxygenase.
J Clin Invest
2007
117
1096
1106
56
Cook
CH
Bickerstaff
AA
Wang
JJ
et al
Spontaneous renal allograft acceptance associated with “regulatory” dendritic cells and IDO.
J Immunol
2008
180
3103
3112
57
Hainz
U
Jürgens
B
Wekerle
T
Seidel
MG
Heitger
A
Indoleamine 2,3-dioxygenase in hematopoietic stem cell transplantation.
Curr Drug Metab
2007
8
267
272
58
Jasperson
LK
Bucher
C
Panoskaltsis-Mortari
A
et al
Indoleamine 2,3-dioxygenase is a critical regulator of acute graft-versus-host disease lethality.
Blood
2008
111
3257
3265
59
Le Blanc
K
Frassoni
F
Ball
L
et al
Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study.
Lancet
2008
371
1579
1586
60
Dunn
GP
Bruce
AT
Ikeda
H
Old
LJ
Schreiber
RD
Cancer immunoediting: from immunosurveillance to tumor escape.
Nat Immunol
2002
3
991
998
61
Okamoto
A
Nikaido
T
Ochiai
K
et al
Indoleamine 2,3-dioxygenase serves as a marker of poor prognosis in gene expression profiles of serous ovarian cancer cells.
Clin Cancer Res
2005
11
6030
6039
62
Ino
K
Yoshida
N
Kajiyama
H
et al
Indoleamine 2,3-dioxygenase is a novel prognostic indicator for endometrial cancer.
Br J Cancer
2006
95
1555
1561
63
Brandacher
G
Perathoner
A
Ladurner
R
et al
Prognostic value of indoleamine 2,3-dioxygenase expression in colorectal cancer: effect on tumor-infiltrating T cells.
Clin Cancer Res
2006
12
1144
1151
64
Muller
AJ
DuHadaway
JB
Donover
PS
Sutanto-Ward
E
Prendergast
GC
Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy.
Nat Med
2005
11
312
319
65
Passegué
E
Jamieson
CH
Ailles
LE
Weissman
IL
Normal and leukemic hematopoiesis: are leukemias a stem cell disorder or a reacquisition of stem cell characteristics?
Proc Natl Acad Sci U S A
2003
100
11842
11849
66
Dazzi
F
Fozza
C
Disease relapse after haematopoietic stem cell transplantation: risk factors and treatment.
Best Pract Res Clin Haematol
2007
20
311
327
67
Molldrem
JJ
Lee
PP
Wang
C
et al
Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia.
Nat Med
2000
6
1018
1023
68
Rezvani
K
Yong
AS
Mielke
S
et al
Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies.
Blood
2008
111
236
242
69
Buggins
AG
Milojkovic
D
Arno
MJ
et al
Microenvironment produced by acute myeloid leukemia cells prevents T cell activation and proliferation by inhibition of NF-κB, c-Myc, and pRb pathways.
J Immunol
2001
167
6021
6030
70
Buggins
AG
Lea
N
Gäken
J
et al
Effect of costimulation and the microenvironment on antigen presentation by leukemic cells.
Blood
1999
94
3479
3490
71
Molldrem
JJ
Lee
PP
Kant
S
et al
Chronic myelogenous leukemia shapes host immunity by selective deletion of high-avidity leukemia-specific T cells.
J Clin Invest
2003
111
639
647
72
Corm
S
Berthon
C
Imbenotte
M
et al
Indoleamine 2,3-dioxygenase activity of acute myeloid leukemia cells can be measured from patients' sera by HPLC and is inducible by IFN-gamma.
Leuk Res
2009
33
490
494
73
Sun
JX
Zhang
WG
Chen
YX
et al
Indoleamine 2, 3-dioxygenase expression in cells of human acute monocyte leukemia (M(5)) and acute lymphocyte leukemia and therapeutic effect of its inhibitor 1-methyl tryptophan.
Zhongguo Shi Yan Xue Ye Xue Za Zhi
2007
15
478
482
74
Tang
XQ
Zhao
ZG
Wang
HX
Li
QB
J
Zou
P
Indoleamine 2, 3-dioxygenase activity in acute myeloid leukemia cells contributing to tumor immune escape.
Zhongguo Shi Yan Xue Ye Xue Za Zhi
2006
14
539
542
75
Chamuleau
M
van de Loosdrecht
AA
Hess
CJ
et al
High INDO (indoleamine 2,3-dioxygenase) mRNA level in blasts of acute myeloid leukemic patients predicts poor clinical outcome.
Haematologica
2008
93
1894
1898
76
Woo
EY
Chu
CS
Goletz
TJ
et al
Regulatory CD4(+)CD25(+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer.
Cancer Res
2001
61
4766
4772
77
Curiel
TJ
Coukos
G
Zou
L
et al
Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival.
Nat Med
2004
10
942
949
78
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
612
79
Marshall
NA
Christie
LE
Munro
LR
et al
Immunosuppressive regulatory T cells are abundant in the reactive lymphocytes of Hodgkin lymphoma.
Blood
2004
103
1755
1762
80
Yang
ZZ
Novak
AJ
Stenson
MJ
Witzig
TE
Ansell
SM
Intratumoral CD4+CD25+ regulatory T-cell-mediated suppression of infiltrating CD4+ T cells in B-cell non-Hodgkin lymphoma.
Blood
2006
107
3639
3646
81
Beyer
M
Kochanek
M
Darabi
K
et al
Reduced frequencies and suppressive function of CD4+CD25hi regulatory T cells in patients with chronic lymphocytic leukemia after therapy with fludarabine.
Blood
2005
106
2018
2025
82
Wang
X
Zheng
J
Liu
J
et al
Increased population of CD4(+)CD25(high), regulatory T cells with their higher apoptotic and proliferating status in peripheral blood of acute myeloid leukemia patients.
Eur J Haematol
2005
75
468
476
83
Beyer
M
Kochanek
M
Giese
T
et al
In vivo peripheral expansion of naive CD4+CD25high FoxP3+ regulatory T cells in patients with multiple myeloma.
Blood
2006
107
3940
3949
84
Kordasti
SY
Ingram
W
Hayden
J
et al
CD4+CD25high Foxp3+ regulatory T cells in myelodysplastic syndrome (MDS).
Blood
2007
110
847
850
85
Mittal
S
Marshall
NA
Duncan
L
Culligan
DJ
Barker
RN
Vickers
MA
Local and systemic induction of CD4+CD25+ regulatory T-cell population by non-Hodgkin lymphoma.
Blood
2008
111
5359
5370
86
Valzasina
B
Piconese
S
Guiducci
C
Colombo
MP
Tumor-induced expansion of regulatory T cells by conversion of CD4+CD25- lymphocytes is thymus and proliferation independent.
Cancer Res
2006
66
4488
4495
87
Chen
W
Jin
W
Hardegen
N
et al
Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-β induction of transcription factor Foxp3.
J Exp Med
2003
198
1875
1886
88
Bronte
V
Zanovello
P
Regulation of immune responses by L-arginine metabolism.
Nat Rev Immunol
2005
5
641
654
89
Novitskiy
SV
Ryzhov
S
Zaynagetdinov
R
et al
Adenosine receptors in regulation of dendritic cell differentiation and function.
Blood
2008
112
1822
1831
90
Casares
N
Pequignot
MO
Tesniere
A
et al
Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death.
J Exp Med
2005
202
1691
1701
91
Gattinoni
L
Powell
DJ
Jr.
Rosenberg
SA
Restifo
NP
Adoptive immunotherapy for cancer: building on success.
Nat Rev Immunol
2006
6
383
393
92
Hou
DY
Muller
AJ
Sharma
MD
et al
Inhibition of indoleamine 2,3-dioxygenase in dendritic cells by stereoisomers of 1-methyl-tryptophan correlates with antitumor responses.
Cancer Res
2007
67
792
801
93
Muller
AJ
Malachowski
WP
Prendergast
GC
Indoleamine 2,3-dioxygenase in cancer: targeting pathological immune tolerance with small-molecule inhibitors.
Expert Opin Ther Targets
2005
9
831
849
94
Banerjee
T
Duhadaway
JB
Gaspari
P
et al
A key in vivo antitumor mechanism of action of natural product-based brassinins is inhibition of indoleamine 2,3-dioxygenase.
Oncogene
2008
27
2851
2857
Sign in via your Institution