1
|
Lemos H, Huang L, Prendergast GC, Mellor AL. Immune control by amino acid catabolism during tumorigenesis and therapy. Nat Rev Cancer 2019; 19:162-175. [PMID: 30696923 DOI: 10.1038/s41568-019-0106-z] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Immune checkpoints arise from physiological changes during tumorigenesis that reprogramme inflammatory, immunological and metabolic processes in malignant lesions and local lymphoid tissues, which constitute the immunological tumour microenvironment (TME). Improving clinical responses to immune checkpoint blockade will require deeper understanding of factors that impact local immune balance in the TME. Elevated catabolism of the amino acids tryptophan (Trp) and arginine (Arg) is a common TME hallmark at clinical presentation of cancer. Cells catabolizing Trp and Arg suppress effector T cells and stabilize regulatory T cells to suppress immunity in chronic inflammatory diseases of clinical importance, including cancers. Processes that induce Trp and Arg catabolism in the TME remain incompletely defined. Indoleamine 2,3 dioxygenase (IDO) and arginase 1 (ARG1), which catabolize Trp and Arg, respectively, respond to inflammatory cues including interferons and transforming growth factor-β (TGFβ) cytokines. Dying cells generate inflammatory signals including DNA, which is sensed to stimulate the production of type I interferons via the stimulator of interferon genes (STING) adaptor. Thus, dying cells help establish local conditions that suppress antitumour immunity to promote tumorigenesis. Here, we review evidence that Trp and Arg catabolism contributes to inflammatory processes that promote tumorigenesis, impede immune responses to therapy and might promote neurological comorbidities associated with cancer.
Collapse
Affiliation(s)
- Henrique Lemos
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-upon-Tyne, UK
| | - Lei Huang
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-upon-Tyne, UK
| | | | - Andrew L Mellor
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-upon-Tyne, UK.
| |
Collapse
|
2
|
Fernández A, Oliver L, Alvarez R, Fernández LE, Mesa C. GM3-containing nanoparticles in immunosuppressed hosts: Effect on myeloid-derived suppressor cells. World J Immunol 2014; 4:98-106. [DOI: 10.5411/wji.v4.i2.98] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 05/27/2014] [Accepted: 06/27/2014] [Indexed: 02/05/2023] Open
Abstract
Cancer vaccines to date have not broadly achieved a significant impact on the overall survival of patients. The negative effect on the immune system of the tumor itself and conventional anti-tumor treatments such as chemotherapy is, undoubtedly, a key reason for these disappointing results. Myeloid-derived suppressor cells (MDSCs) are considered a central node of the immunosuppressive network associated with tumors. These cells inhibit the effector function of natural killer and CD8+ T cells, expand regulatory T cells and can differentiate into tumor-associated macrophages within the tumor microenvironment. Thus, overcoming the suppressive effects of MDSCs is likely to be critical for cancer immunotherapy to generate effective anti-tumor immune responses. However, the capacity of cancer vaccines and particularly their adjuvants to overcome this inhibitory population has not been well characterized. Very small size proteoliposomes (VSSP) is a nanoparticulated adjuvant specifically designed to be formulated with vaccines used in the treatment of immunocompromised patients. This adjuvant contains immunostimulatory bacterial signals together with GM3 ganglioside. VSSP promotes dendritic cell maturation, antigen cross-presentation to CD8+ T cells, Th1 polarization, and enhances CD8+ T cell response in tumor-free mice. Currently, four cancer vaccines using VSSP as the adjuvant are in Phase I and II clinical trials. In this review, we summarize our work characterizing the unique ability of VSSP to stimulate antigen-specific CD8+ T cell responses in two immunocompromised scenarios; in tumor-bearing mice and during chemotherapy-induced leukopenia. Particular emphasis has been placed on the interaction of these nanoparticles with MDSCs, as well as comparison with other cancer vaccine adjuvants currently in preclinical or clinical studies.
Collapse
|
3
|
Upreti D, Pathak A, Kung SKP. Lentiviral vector-based therapy in head and neck cancer (Review). Oncol Lett 2013; 7:3-9. [PMID: 24348811 PMCID: PMC3861563 DOI: 10.3892/ol.2013.1652] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 09/17/2013] [Indexed: 12/30/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common neoplasm worldwide. Despite advances in multimodality treatments involving surgery, radiation and chemotherapy, the five-year survival rate has remained at ~50% for the past 35 years. Therefore, the early detection of recurrent or persistent disease is extremely important. Replication-incompetent HIV-1-based lentiviral vectors have emerged as powerful and safe tools for gene delivery. Commonly, HNSCC is a locoregional disease that presents at or close to the body surface. Thus, HNSCC is amendable to intratumoral injections of gene therapy vectors aimed at correcting defects associated with tumor suppressor genes to induce the direct cytotoxicity of cancer cells or immune modulation to promote antitumor immunity. Current investigations analyzing HNSCC gene mutations and stem cell markers and the cancer immunoediting concept are creating exciting therapeutic opportunities for lentiviral and other gene transfer vectors. The present review reports specific examples of the current applications of lentiviral vectors in HNSCC.
Collapse
Affiliation(s)
- Deepak Upreti
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Alok Pathak
- Department of Surgery, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Sam K P Kung
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
4
|
Oliver L, Fernández A, Raymond J, López-Requena A, Fernández LE, Mesa C. Very small size proteoliposomes derived from Neisseria meningitidis: an effective adjuvant for antigen-specific cytotoxic T lymphocyte response stimulation under leukopenic conditions. Vaccine 2012; 30:2963-72. [PMID: 22391399 DOI: 10.1016/j.vaccine.2012.02.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 02/16/2012] [Accepted: 02/19/2012] [Indexed: 10/28/2022]
Abstract
Leukopenia is a severe condition resulting from both pathological processes and some treatments, like chemotherapy in cancer patients. However, the activation of the patient immune system is required for the success of immunotherapeutic strategies, as cancer vaccines. In this regard, leukopenia constitutes a major hurdle to overcome, mainly due to the impairment of cytotoxic T lymphocyte (CTL) responses. Adjuvants are basic components of vaccine formulations, which might be useful to stimulate immunity under this immunosuppressed condition. To this aim, we tested the capacity of a novel nanoparticulated complex, very small size proteoliposomes (VSSP), to promote CTL even in a leukopenic scenario. Noteworthy, we observed that a VSSP-based OVA vaccine induced a normal antigen-specific CTL response in mice rendered leukopenia by the administration of high doses of the chemotherapeutic agent cyclophosphamide (CY), while under the same conditions the OVA antigen formulated in the TLR-3 agonist polyinosinic-polycytidylic acid (P(I:C)) was ineffective. Moreover, an appropriate combination of VSSP with the P(I:C) vaccine was able to restore the CD8(+) T cell effector function in leukopenic mice. VSSP induced not only a faster repopulation of immune cells in CY-receiving animals, but also enhanced the recovery of memory T lymphocytes and myeloid dendritic cells (DCs) while simultaneously abrogated the immunosuppressive capacity of myeloid-derived suppressor cells (MDSCs). Our results suggest that VSSP could be a particularly suitable immunomodulator to be used in CTL-promoting active immunotherapy strategies operating in severe immune compromised scenarios.
Collapse
Affiliation(s)
- Liliana Oliver
- Center of Molecular Immunology, Atabey, Playa, Havana 11600, Cuba
| | | | | | | | | | | |
Collapse
|
5
|
Johnson BA, Baban B, Mellor AL. Targeting the immunoregulatory indoleamine 2,3 dioxygenase pathway in immunotherapy. Immunotherapy 2011; 1:645-61. [PMID: 20161103 DOI: 10.2217/imt.09.21] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Natural immune tolerance is a formidable barrier to successful immunotherapy to treat established cancers and chronic infections. Conversely, creating robust immune tolerance via immunotherapy is the major goal in treating autoimmune and allergic diseases, and enhancing survival of transplanted organs and tissues. In this review, we focus on a natural mechanism that creates local T-cell tolerance in many clinically relevant settings of chronic inflammation involving expression of the cytosolic enzyme indoleamine 2,3-dioxygenase (IDO) by specialized subsets of dendritic cells. IDO-expressing dendritic cells suppress antigen-specific T-cell responses directly, and induce bystander suppression by activating regulatory T cells. Thus, manipulating IDO is a promising strategy to treat a range of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Burles A Johnson
- Immunotherapy Center & Department of Medicine, Medical College of Georgia, Augusta, GA 30912, USA.
| | | | | |
Collapse
|
6
|
Huang L, Baban B, Johnson BA, Mellor AL. Dendritic cells, indoleamine 2,3 dioxygenase and acquired immune privilege. Int Rev Immunol 2010; 29:133-55. [PMID: 20367139 DOI: 10.3109/08830180903349669] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Dendritic cells (DCs) are specialized to stimulate T cell immunity. Paradoxically, some DCs suppress T cell responses and activate regulatory T cells. In this review, we focus on a potent counter-regulatory pathway mediated by plasmacytoid DCs (pDCs) expressing the immunosuppressive enzyme indoleamine 2,3 dioxygenase (IDO). IDO-expressing pDCs inhibit effector T cell responses, activate regulatory T cells, and attenuate pro-inflammatory responses in settings of chronic inflammation that manifest in clinical syndromes, such as infectious, allergic, and autoimmune diseases; cancer; and transplantation. Thus, IDO-expressing pDCs create immune privilege and provide novel opportunities to improve immunotherapy in multiple disease syndromes.
Collapse
Affiliation(s)
- Lei Huang
- Immunotherapy Center and Department of Radiology, Medical College of Georgia, Augusta, GA, USA
| | | | | | | |
Collapse
|
7
|
Immunotherapy in head and neck cancer: current practice and future possibilities. The Journal of Laryngology & Otology 2008; 123:19-28. [PMID: 18761763 DOI: 10.1017/s0022215108003356] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The survival of patients with head and neck squamous cell carcinoma has changed little over the last 30 years. However, with recent advances in the fields of cellular and molecular immunology, there is renewed optimism with regards to the development of novel methods of early diagnosis, prognosis estimation and treatment improvement for patients with head and neck squamous cell carcinoma. Here, we present a critical review of the recent advances in tumour immunology, and of the current efforts to apply new immunotherapeutic techniques in the treatment of head and neck squamous cell carcinoma.
Collapse
|
8
|
Devarapu SK, Sharma SC, Das SN. Triggering of T cell-mediated immune responses by allogenic tumor cell vaccine in patients with oral cancer. Immunopharmacol Immunotoxicol 2006; 28:387-95. [PMID: 16997788 DOI: 10.1080/08923970600927348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We investigated the immunomodulatory activity of allogenic whole tumor cell vaccine in oral cancer patients in vitro by two-color flow cytometry. Vaccine treatment significantly increased the expression of CD69 and HLA-DR in CD3+ T-cell subsets. The frequency of Interferon-gamma and Interleukin (IL)-2 expressing CD4+/CD8+ T-cell subsets was significantly higher with a concomitant reduction in IL-4 and IL-10 expressing T-cells in the vaccine treated group as compared with the untreated controls. Vaccine treatment significantly increased T-cell receptor (TCR), Vbeta3, Vbeta5 and Vbeta8 usage. The results indicate that the allogenic whole tumor cell vaccine is able to trigger T-cell mediated immunity in patients with intraoral squamous cell carcinoma.
Collapse
MESH Headings
- Adult
- Aged
- Antigens, CD/analysis
- Antigens, Differentiation, T-Lymphocyte/analysis
- CD3 Complex/analysis
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Female
- Flow Cytometry/methods
- HLA-DR Antigens/analysis
- Humans
- Interferon-gamma/analysis
- Interleukin-10/analysis
- Interleukin-2/analysis
- Interleukin-4/analysis
- Interleukins/analysis
- Interleukins/classification
- Jurkat Cells
- Lectins, C-Type
- Male
- Middle Aged
- Mouth Neoplasms/drug therapy
- Mouth Neoplasms/immunology
- Mouth Neoplasms/metabolism
- Neoplasms, Squamous Cell/drug therapy
- Neoplasms, Squamous Cell/immunology
- Neoplasms, Squamous Cell/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/analysis
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Satish K Devarapu
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | | |
Collapse
|
9
|
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. THE JOURNAL OF IMMUNOLOGY 2004; 172:4100-10. [PMID: 15034022 DOI: 10.4049/jimmunol.172.7.4100] [Citation(s) in RCA: 351] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Human monocyte-derived dendritic cells (DCs) are capable of expressing the tryptophan-degrading enzyme indoleamine 2,3-dioxygenase (IDO), which allows them to suppress Ag-driven proliferation of T cells in vitro. In DCs that express IDO, the activity of the enzyme is tightly regulated, with the protein being constitutively expressed, but functional activity requiring an additional set of triggering signals supplied during Ag presentation. We now show that triggering of functional IDO obligately requires ligation of B7-1/B7-2 molecules on the DCs by CTLA4/CD28 expressed on T cells. When this interaction was disrupted, IDO remained in the inactive state, and the DCs were unable to inhibit T cell proliferation. Inhibition could be fully restored by direct Ab-mediated cross-linking of B7-1/B7-2. Although both CD4(+) and CD8(+) T cells were susceptible to inhibition once IDO was induced, the ability to trigger functionally active IDO was strictly confined to the CD4(+) subset. Thus, the ability of CD4(+) T cells to induce IDO activity in DCs allowed the CD4(+) population to dominantly inhibit proliferation of the CD8(+) population via the bridge of a conditioned DC. We hypothesize that IDO activation via engagement of B7-1/B7-2 molecules on DCs, specifically, engagement by CTLA4 expressed on regulatory CD4(+) T cells, may function as a physiologic regulator of T cell responses in vivo.
Collapse
Affiliation(s)
- David H Munn
- Institute of Molecular Medicine and Genetics and Departments of Pediatrics, Medical College of Georgia, Augusta, GA 30912, USA.
| | | | | |
Collapse
|
10
|
Misra N, Bayry J, Lacroix-Desmazes S, Kazatchkine MD, Kaveri SV. Cutting edge: human CD4+CD25+ T cells restrain the maturation and antigen-presenting function of dendritic cells. THE JOURNAL OF IMMUNOLOGY 2004; 172:4676-80. [PMID: 15067041 DOI: 10.4049/jimmunol.172.8.4676] [Citation(s) in RCA: 352] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The characteristics and functions of CD4(+)CD25(+) regulatory cells have been well defined in murine and human systems. However, the interaction between CD4(+)CD25(+) T cells and dendritic cells (DC) remains unclear. In this study, we examined the effect of human CD4(+)CD25(+) T cells on maturation and function of monocyte-derived DC. We show that regulatory T cells render the DC inefficient as APCs despite prestimulation with CD40 ligand. This effect was marginally reverted by neutralizing Abs to TGF-beta. There was an increased IL-10 secretion and reduced expression of costimulatory molecules in DC. Thus, in addition to direct suppressor effect on CD4(+) T cells, regulatory T cells may modulate the immune response through DC.
Collapse
Affiliation(s)
- Namita Misra
- Institut National de la Santé et de la Recherche Médicale, Unité 430, and Université Pierre et Marie Curie, Centre de Recherches Biomedicales des Cordeliers, Paris, France
| | | | | | | | | |
Collapse
|
11
|
Zibert A, Balzer S, Souquet M, Quang TH, Paris-Scholz C, Roskrow M, Dilloo D. CCL3/MIP-1αIs a Potent Immunostimulator When Coexpressed with Interleukin-2 or Granulocyte-Macrophage Colony-Stimulating Factor in a Leukemia/Lymphoma Vaccine. Hum Gene Ther 2004; 15:21-34. [PMID: 14965375 DOI: 10.1089/10430340460732436] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Chemokines orchestrate trafficking of immune effector cells during inflammation. Here we demonstrate that chemokines also serve to potentiate effector cell-mediated antineoplastic immune responses in vaccination strategies. As a critical mediator of inflammation, macrophage inflammatory protein 1alpha (CCL3/MIP-1alpha) attracts and stimulates both antigen-presenting and cytotoxic cells. In the A20 leukemia/lymphoma vaccine model, we explored the efficacy of MIP-1alpha in combination with interleukin-2 (IL-2) or granulocyte-macrophage colony-stimulating factor (GM-CSF). After subcutaneous injection of the MIP-1alpha + IL-2 or MIP-1alpha + GM-CSF combination vaccine, focal but pronounced infiltrates of CD4+ and CD8+ T cells were observed at the vaccination sites. In mice with preestablished leukemia/lymphoma, survival is significantly improved in animals treated with MIP-1alpha + GM-CSF- and MIP-1alpha + IL-2-secreting vaccines. Protection is superior in the MIP-1alpha + GM-CSF group, with the effects of MIP-1alpha and GM-CSF being synergistic. In contrast, suppression of lymphoblast proliferation by single-immunogen vaccines secreting MIP-1alpha, GM-CSF, or IL-2 alone does not translate to improved survival. The systemic protective effects afforded by the MIP-1alpha + IL-2 or MIP-1alpha + GM-CSF combination are mediated by different effector cell populations. In the MIP-1alpha + IL-2 group, antineoplastic defense is mediated by CD8+ T and NK cells, whereas in the MIP-1alpha + GM-CSF group CD4+ T cells are involved in addition to CD8+ cytotoxic T cells, underscoring that T cell help is critical for long-term protection. Thus combination of MIP-1alpha with different cytokines recruits different sets of effector cells into a potent antineoplastic immune response.
Collapse
Affiliation(s)
- Andree Zibert
- Clinic for Pediatric Oncology, Hematology, and Immunology, University Hospital Dusseldorf, D-40225 Dusseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
12
|
Kacani L, Wurm M, Schennach H, Braun I, Andrle J, Sprinzl GM. Immunosuppressive effects of soluble factors secreted by head and neck squamous cell carcinoma on dendritic cells and T lymphocytes. Oral Oncol 2003; 39:672-9. [PMID: 12907206 DOI: 10.1016/s1368-8375(03)00076-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent observations suggest that the inability of the immune system to mount an effective immune response against head and neck squamous cell carcinoma (HNSCC) could be a result of the immunosuppression mediated through soluble factors that are secreted by tumour cells. Therefore, we investigated the effects of conditioned medium obtained from cultures of HNSCC cell lines (HNSCC-CM) on the function of dendritic cells (DC) and T cell immune response. In our study, we could not observe any inhibitory effect of HNSCC-CM on the maturation and the cytokine secretion pattern of DC. On the contrary, HNSCC-CM from two of three cell lines consistently decreased the quantity of IFN-gamma- and IL-4-secreting T cells upon restimulation in vitro. In conclusion, our data suggest that soluble factors secreted by HNSCC cells directly inhibit the function of effector T cells, rather than impeding the process of antigen presentation.
Collapse
Affiliation(s)
- Laco Kacani
- Department of Otorhinolaryngology, University Hospital Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
13
|
Mellor AL, Baban B, Chandler P, Marshall B, Jhaver K, Hansen A, Koni PA, Iwashima M, Munn DH. Cutting edge: induced indoleamine 2,3 dioxygenase expression in dendritic cell subsets suppresses T cell clonal expansion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:1652-5. [PMID: 12902462 DOI: 10.4049/jimmunol.171.4.1652] [Citation(s) in RCA: 359] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In mice, immunoregulatory APCs express the dendritic cell (DC) marker CD11c, and one or more distinctive markers (CD8alpha, B220, DX5). In this study, we show that expression of the tryptophan-degrading enzyme indoleamine 2,3 dioxygenase (IDO) is selectively induced in specific splenic DC subsets when mice were exposed to the synthetic immunomodulatory reagent CTLA4-Ig. CTLA4-Ig did not induce IDO expression in macrophages or lymphoid cells. Induction of IDO completely blocked clonal expansion of T cells from TCR transgenic mice following adoptive transfer, whereas CTLA4-Ig treatment did not block T cell clonal expansion in IDO-deficient recipients. Thus, IDO expression is an inducible feature of specific subsets of DCs, and provides a potential mechanistic explanation for their T cell regulatory properties.
Collapse
Affiliation(s)
- Andrew L Mellor
- Department of Medicine, Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, GA 30912, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Affiliation(s)
- Nancy Hogg
- Cancer Research UK London Research Institute, London, UK.
| |
Collapse
|
15
|
Mellor AL, Munn DH. Tryptophan catabolism and regulation of adaptive immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5809-13. [PMID: 12794104 DOI: 10.4049/jimmunol.170.12.5809] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Andrew L Mellor
- Department of Medicine, Medical College of Georgia, Augusta GA 30912, USA
| | | |
Collapse
|