1
|
Wang A, Wang Y, Liang R, Li B, Pan F. Improving regulatory T cell-based therapy: insights into post-translational modification regulation. J Genet Genomics 2024:S1673-8527(24)00252-2. [PMID: 39357622 DOI: 10.1016/j.jgg.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Regulatory T (Treg) cells are pivotal for maintaining immune homeostasis and play essential roles in various diseases, such as autoimmune diseases, graft-versus-host disease (GVHD), tumors, and infectious diseases. Treg cells exert suppressive function via distinct mechanisms including inhibitory cytokines, granzyme or perforin-mediated cytolysis, metabolic disruption, and suppression of dendritic cells. Forkhead Box P3 (FOXP3), the characteristic transcription factor, is essential for Treg cell function and plasticity. Cumulative evidence has demonstrated that FOXP3 activity and Treg cell function are modulated by a variety of post-translational modifications (PTMs), including ubiquitination, acetylation, phosphorylation, methylation, glycosylation, poly(ADP-ribosyl)ation, and uncharacterized modifications. This review describes Treg cell suppressive mechanisms and summarizes the current evidence on PTM regulation of FOXP3 and Treg cell function. Understanding the regulatory role of PTMs in Treg cell plasticity and function will be helpful in designing therapeutic strategies for autoimmune diseases, GVHD, tumors, and infectious diseases.
Collapse
Affiliation(s)
- Aiting Wang
- Center for Cancer Immunology Research, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| | - Yanwen Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rui Liang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Fan Pan
- Center for Cancer Immunology Research, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
2
|
Rodriguez S, Alizadeh M, Lamaison C, Saintamand A, Monvoisin C, Jean R, Deleurme L, Martin-Subero JI, Pangault C, Cogné M, Amé-Thomas P, Tarte K. Follicular lymphoma regulatory T-cell origin and function. Front Immunol 2024; 15:1391404. [PMID: 38799444 PMCID: PMC11116630 DOI: 10.3389/fimmu.2024.1391404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Follicular Lymphoma (FL) results from the malignant transformation of germinal center (GC) B cells. FL B cells display recurrent and diverse genetic alterations, some of them favoring their direct interaction with their cell microenvironment, including follicular helper T cells (Tfh). Although FL-Tfh key role is well-documented, the impact of their regulatory counterpart, the follicular regulatory T cell (Tfr) compartment, is still sparse. Methods The aim of this study was to characterize FL-Tfr phenotype by cytometry, gene expression profile, FL-Tfr origin by transcriptomic analysis, and functionality by in vitro assays. Results CD4+CXCR5+CD25hiICOS+ FL-Tfr displayed a regulatory program that is close to classical regulatory T cell (Treg) program, at the transcriptomic and methylome levels. Accordingly, Tfr imprinting stigmata were found on FL-Tfh and FL-B cells, compared to their physiological counterparts. In addition, FL-Tfr co-culture with autologous FL-Tfh or cytotoxic FL-CD8+ T cells inhibited their proliferation in vitro. Finally, although FL-Tfr shared many characteristics with Treg, TCR sequencing analyses demonstrated that part of them derived from precursors shared with FL-Tfh. Discussion Altogether, these findings uncover the role and origin of a Tfr subset in FL niche and may be useful for lymphomagenesis knowledge and therapeutic management.
Collapse
Affiliation(s)
- Stéphane Rodriguez
- Unité Mixte de Recherche (UMR)1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
| | - Mehdi Alizadeh
- Service Recherche, Etablissement Français du Sang, Rennes, France
| | - Claire Lamaison
- Unité Mixte de Recherche (UMR)1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
| | - Alexis Saintamand
- Unité Mixte de Recherche (UMR)1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
| | - Céline Monvoisin
- Unité Mixte de Recherche (UMR)1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
| | - Rachel Jean
- Unité Mixte de Recherche (UMR)1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
- Pôle Biologie, Centre Hospitalier Universitaire, Rennes, France
| | - Laurent Deleurme
- Unité Mixte de Recherche (UMR)1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
- Univ Rennes, CNRS, INSERM, BIOSIT (BIOlogie, Santé, Innovation Technologique de Rennes) – Unité Mixte de Service 34 80, Rennes, France
| | - Jose Ignacio Martin-Subero
- Departamento de Anatomía Patológica, Farmacología y Microbiología, Universitat de Barcelona, Barcelona, Spain
| | - Céline Pangault
- Unité Mixte de Recherche (UMR)1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
- Pôle Biologie, Centre Hospitalier Universitaire, Rennes, France
| | - Michel Cogné
- Unité Mixte de Recherche (UMR)1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
| | - Patricia Amé-Thomas
- Unité Mixte de Recherche (UMR)1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
- Pôle Biologie, Centre Hospitalier Universitaire, Rennes, France
| | - Karin Tarte
- Unité Mixte de Recherche (UMR)1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
- Suivi Immunologique des Thérapeutiques Innovantes (SITI) Laboratory, Centre Hospitalier Universitaire Rennes, Etablissement Français du Sang Bretagne, Rennes, France
| |
Collapse
|
3
|
Ashenafi S, Brighenti S. Reinventing the human tuberculosis (TB) granuloma: Learning from the cancer field. Front Immunol 2022; 13:1059725. [PMID: 36591229 PMCID: PMC9797505 DOI: 10.3389/fimmu.2022.1059725] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Tuberculosis (TB) remains one of the deadliest infectious diseases in the world and every 20 seconds a person dies from TB. An important attribute of human TB is induction of a granulomatous inflammation that creates a dynamic range of local microenvironments in infected organs, where the immune responses may be considerably different compared to the systemic circulation. New and improved technologies for in situ quantification and multimodal imaging of mRNA transcripts and protein expression at the single-cell level have enabled significantly improved insights into the local TB granuloma microenvironment. Here, we review the most recent data on regulation of immunity in the TB granuloma with an enhanced focus on selected in situ studies that enable spatial mapping of immune cell phenotypes and functions. We take advantage of the conceptual framework of the cancer-immunity cycle to speculate how local T cell responses may be enhanced in the granuloma microenvironment at the site of Mycobacterium tuberculosis infection. This includes an exploratory definition of "hot", immune-inflamed, and "cold", immune-excluded TB granulomas that does not refer to the level of bacterial replication or metabolic activity, but to the relative infiltration of T cells into the infected lesions. Finally, we reflect on the current knowledge and controversy related to reactivation of active TB in cancer patients treated with immune checkpoint inhibitors such as PD-1/PD-L1 and CTLA-4. An understanding of the underlying mechanisms involved in the induction and maintenance or disruption of immunoregulation in the TB granuloma microenvironment may provide new avenues for host-directed therapies that can support standard antibiotic treatment of persistent TB disease.
Collapse
Affiliation(s)
- Senait Ashenafi
- Department of Medicine Huddinge, Center for Infectious Medicine (CIM), Karolinska Institutet, ANA Futura, Huddinge, Sweden,Department of Pathology, School of Medicine, College of Health Sciences, Tikur Anbessa Specialized Hospital and Addis Ababa University, Addis Ababa, Ethiopia
| | - Susanna Brighenti
- Department of Medicine Huddinge, Center for Infectious Medicine (CIM), Karolinska Institutet, ANA Futura, Huddinge, Sweden,*Correspondence: Susanna Brighenti,
| |
Collapse
|
4
|
Kumar E, Pickard L, Okosun J. Pathogenesis of follicular lymphoma: genetics to the microenvironment to clinical translation. Br J Haematol 2021; 194:810-821. [PMID: 33694181 DOI: 10.1111/bjh.17383] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/08/2021] [Indexed: 01/10/2023]
Abstract
Follicular lymphoma (FL) represents a heterogeneous disease both clinically and biologically. The pathognomonic t(14;18) translocation can no longer be thought of as the primary genetic driver, with increasing recognition of the biological relevance of recurrent genetic alterations in epigenetic regulators that now feature as a pivotal hallmark of this lymphoma subtype. Furthermore, sequencing studies have provided a near complete catalogue of additional genetic aberrations. Longitudinal and spatial genetic studies add an additional layer to the biological heterogeneity, providing preliminary molecular insights into high-risk phenotypes such as early progressors and transformation, and also supporting evidence for the existence of persisting re-populating cells that act as lymphoma reservoirs and harbingers for FL recurrence. Simultaneously, understanding of the tumour microenvironmental cues promoting lymphomagenesis and disease progression continue to broaden. More recently, studies are beginning to unravel the convergence and co-operation between the genetics, epigenetics and microenvironment. There is a pressing need to marry biology with therapeutics, especially with the burgeoning treatment landscape in FL, to aid in optimising patient selection and guiding the 'right drug to the right patient'.
Collapse
Affiliation(s)
- Emil Kumar
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Lucy Pickard
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jessica Okosun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
5
|
Scott EN, Gocher AM, Workman CJ, Vignali DAA. Regulatory T Cells: Barriers of Immune Infiltration Into the Tumor Microenvironment. Front Immunol 2021; 12:702726. [PMID: 34177968 PMCID: PMC8222776 DOI: 10.3389/fimmu.2021.702726] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
Regulatory T cells (Tregs) are key immunosuppressive cells that promote tumor growth by hindering the effector immune response. Tregs utilize multiple suppressive mechanisms to inhibit pro-inflammatory responses within the tumor microenvironment (TME) by inhibition of effector function and immune cell migration, secretion of inhibitory cytokines, metabolic disruption and promotion of metastasis. In turn, Tregs are being targeted in the clinic either alone or in combination with other immunotherapies, in efforts to overcome the immunosuppressive TME and increase anti-tumor effects. However, it is now appreciated that Tregs not only suppress cells intratumorally via direct engagement, but also serve as key interactors in the peritumor, stroma, vasculature and lymphatics to limit anti-tumor immune responses prior to tumor infiltration. We will review the suppressive mechanisms that Tregs utilize to alter immune and non-immune cells outside and within the TME and discuss how these mechanisms collectively allow Tregs to create and promote a physical and biological barrier, resulting in an immune-excluded or limited tumor microenvironment.
Collapse
Affiliation(s)
- Ellen N. Scott
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Angela M. Gocher
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
| | - Creg J. Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dario A. A. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
6
|
The Tumor Microenvironment in Follicular Lymphoma: Its Pro-Malignancy Role with Therapeutic Potential. Int J Mol Sci 2021; 22:ijms22105352. [PMID: 34069564 PMCID: PMC8160856 DOI: 10.3390/ijms22105352] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
In the follicular lymphoma (FL) microenvironment, CXCR5+ICOS+PD1+BCL6+ follicular helper T (Tfh) cells, which closely correlate with FL B cells in neoplastic follicles, play a major role in supporting FL. Interleukin-4 secreted by Tfh cells triggers the upregulation of the lymphocyte chemoattractant CXCL12 in stromal cell precursors, in particular by fibroblastic reticular cells (FRCs). In turn, mesenchymal stem cells (MSCs) can be committed to FRC differentiation in the bone marrow and lymph nodes involved by FL. Noteworthy, MSCs can promote the differentiation of Tfh cells into highly immunosuppressive T-follicular regulatory cells. The tumor suppressor HVEM is highly mutated in FL cells, and its deficiency increases Tfh cell frequency. In contrast, PI3Kδ inhibition impedes the recruitment of Tfh/regulatory T cells and impairs the proliferation of follicular dendritic cells (FDCs) and FDC-induced angiogenesis. Since TIGIT ligands are expressed by FDCs, the immune checkpoint receptor TIGIT plays an important role in tumor-infiltrating T cells. Thus, TIGIT blockade might invigorate cytotoxic T cells in the FL microenvironment. Given their potential to simultaneously reduce the neoplastic B cells, Tfh, and TFR cells could also reinforce the effects of the cytotoxic T cells. This combinatory strategy should be explored as a treatment option to tackle FL.
Collapse
|
7
|
Dong Y, Yang C, Pan F. Post-Translational Regulations of Foxp3 in Treg Cells and Their Therapeutic Applications. Front Immunol 2021; 12:626172. [PMID: 33912156 PMCID: PMC8071870 DOI: 10.3389/fimmu.2021.626172] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Regulatory T (Treg) cells are indispensable for immune homeostasis due to their roles in peripheral tolerance. As the master transcription factor of Treg cells, Forkhead box P3 (Foxp3) strongly regulates Treg function and plasticity. Because of this, considerable research efforts have been directed at elucidating the mechanisms controlling Foxp3 and its co-regulators. Such work is not only advancing our understanding on Treg cell biology, but also uncovering novel targets for clinical manipulation in autoimmune diseases, organ transplantation, and tumor therapies. Recently, many studies have explored the post-translational regulation of Foxp3, which have shown that acetylation, phosphorylation, glycosylation, methylation, and ubiquitination are important for determining Foxp3 function and plasticity. Additionally, some of these targets have been implicated to have great therapeutic values. In this review, we will discuss emerging evidence of post-translational regulations on Foxp3 in Treg cells and their exciting therapeutic applications.
Collapse
Affiliation(s)
- Yi Dong
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Cuiping Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fan Pan
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, China
| |
Collapse
|
8
|
van Bruggen JAC, Martens AWJ, Tonino SH, Kater AP. Overcoming the Hurdles of Autologous T-Cell-Based Therapies in B-Cell Non-Hodgkin Lymphoma. Cancers (Basel) 2020; 12:cancers12123837. [PMID: 33353234 PMCID: PMC7765898 DOI: 10.3390/cancers12123837] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 01/10/2023] Open
Abstract
Simple Summary The activity of novel therapies that utilize patient’s own T-cells to induce remission of B-cell non-Hodgkin lymphoma (B-NHL), including chronic lymphocytic leukemia (CLL), is still suboptimal. In this review, we summarize the clinical efficacy of T-cell-based therapies in B-NHL and provide a biologic rationale for the observed (lack of) responses. We describe and compare the acquired T-cell dysfunctions that occur in the different subtypes of B-NHL. Furthermore, we discuss new insights that could enhance the efficacy of T-cell-based therapies for B-NHL and CLL. Abstract The next frontier towards a cure for B-cell non-Hodgkin lymphomas (B-NHL) is autologous cellular immunotherapy such as immune checkpoint blockade (ICB), bispecific antibodies (BsAbs) and chimeric antigen receptor (CAR) T-cells. While highly successful in various solid malignancies and in aggressive B-cell leukemia, this clinical success is often not matched in B-NHL. T-cell subset skewing, exhaustion, expansion of regulatory T-cell subsets, or other yet to be defined mechanisms may underlie the lack of efficacy of these treatment modalities. In this review, a systematic overview of results from clinical trials is given and is accompanied by reported data on T-cell dysfunction. From these results, we distill the underlying pathways that might be responsible for the observed differences in clinical responses towards autologous T-cell-based cellular immunotherapy modalities between diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), mantle cell lymphoma (MCL), and marginal zone lymphoma (MZL). By integration of the clinical and biological findings, we postulate strategies that might enhance the efficacy of autologous-based cellular immunotherapy for the treatment of B-NHL.
Collapse
Affiliation(s)
- Jaco A. C. van Bruggen
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.A.C.v.B.); (A.W.J.M.); (S.H.T.)
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, 1105 AZ Amsterdam, The Netherlands
| | - Anne W. J. Martens
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.A.C.v.B.); (A.W.J.M.); (S.H.T.)
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, 1105 AZ Amsterdam, The Netherlands
| | - Sanne H. Tonino
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.A.C.v.B.); (A.W.J.M.); (S.H.T.)
- Cancer Center Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, 1105 AZ Amsterdam, The Netherlands
| | - Arnon P. Kater
- Department of Hematology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.A.C.v.B.); (A.W.J.M.); (S.H.T.)
- Cancer Center Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, 1105 AZ Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|
9
|
An integrative microenvironment approach for follicular lymphoma: roles of inflammatory cell subsets and immune-response polymorphisms on disease clinical course. Oncotarget 2020; 11:3153-3173. [PMID: 32913559 PMCID: PMC7443366 DOI: 10.18632/oncotarget.27698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/14/2020] [Indexed: 01/16/2023] Open
Abstract
The study of the tumor microenvironment (TME) in follicular lymphoma (FL) has produced conflicting results due to assessment of limited TME subpopulations, and because of heterogeneous treatments among different cohorts. Also, important genetic determinants of immune response, such as single-nucleotide polymorphisms (SNPs), remain underexplored in this disease. We performed a detailed study of the TME in 169 FL biopsies using immunohistochemistry, encompassing lymphocytes, macrophages, and cytokines. We also genotyped 16 SNPs within key immune-response genes (IL12A, IL2, IL10, TGFB1, TGFBR1, TGFBR2, IL17A, and IL17F) in 159 patients. We tested associations between SNPs, clinicopathological features and TME composition, and proposed survival models in R-CHOP/R-CVP-treated patients. Presence of the IL12A rs568408 "A" allele associated with the follicular pattern of FOXP3+ cells. The IL12A AA haplotype included rs583911 and rs568408 and was an independent predictor of worse survival, together with the follicular patterns of T-cells (FOXP3+ and CD8+) and high IL-17F tumor levels. The patterns of CD3+, CD4+ and CD8+ cells, displayed as a principal component, also associated with survival. Hierarchical clustering of the TME proteins demonstrated a cluster that was associated with worse prognosis (tumors enriched in IL-17A, IL-17F, CD8, PD1, and Ki-67). The survival of FL patients who were treated in the rituximab era shows a strong dependence on TME signals, especially the T-cell infiltration patterns and IL-17F tumor levels. The presence of the AA haplotype of IL12A in the genome of FL patients is an additional prognostic factor that may modulate the composition of T-reg cells in this disease.
Collapse
|
10
|
Gauthier JM, Harrison MS, Krupnick AS, Gelman AE, Kreisel D. The emerging role of regulatory T cells following lung transplantation. Immunol Rev 2019; 292:194-208. [PMID: 31536165 DOI: 10.1111/imr.12801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022]
Abstract
Regulatory T cells (Treg) have proven to be a powerful immunologic force in nearly every organ system and hold therapeutic potential for a wide range of diseases. Insights gained from non-transplant pathologies, such as infection, cancer, and autoimmunity, are now being translated to the field of solid organ transplantation, particularly for livers and kidneys. Recent insights from animal models of lung transplantation have established that Tregs play a vital role in suppressing rejection and facilitating tolerance of lung allografts, and such discoveries are being validated in human studies and preclinical trials. Given that long-term outcomes following lung transplantation remain profoundly limited by chronic rejection, Treg therapy holds the potential to significantly improve patient outcomes and should be aggressively investigated.
Collapse
Affiliation(s)
- Jason M Gauthier
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University, Saint Louis, MO, USA
| | - M Shea Harrison
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University, Saint Louis, MO, USA
| | - Alexander S Krupnick
- Division of Thoracic Surgery, Department of Surgery, University of Virginia, Charlottesville, VA, USA.,Carter Immunology Center, University of Virginia, Charlottesville, VA, USA
| | - Andrew E Gelman
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University, Saint Louis, MO, USA.,Department of Pathology & Immunology, Washington University, Saint Louis, MO, USA
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University, Saint Louis, MO, USA.,Department of Pathology & Immunology, Washington University, Saint Louis, MO, USA
| |
Collapse
|
11
|
Yano H, Andrews LP, Workman CJ, Vignali DAA. Intratumoral regulatory T cells: markers, subsets and their impact on anti-tumor immunity. Immunology 2019; 157:232-247. [PMID: 31087644 DOI: 10.1111/imm.13067] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 12/14/2022] Open
Abstract
Regulatory T (Treg) cells play a crucial role in maintaining self-tolerance and resolution of immune responses by employing multifaceted immunoregulatory mechanisms. However, Treg cells readily infiltrate into the tumor microenvironment (TME) and dampen anti-tumor immune responses, thereby becoming a barrier to effective cancer immunotherapy. There has been a substantial expansion in the development of novel immunotherapies targeting various inhibitory receptors (IRs), such as CTLA4, PD1 and LAG3, but these approaches have mechanistically focused on the elicitation of anti-tumor responses. However, enhanced inflammation in the TME could also play a detrimental role by facilitating the recruitment, stability and function of Treg cells by up-regulating chemokines that promote Treg cell migration, and/or increasing inhibitory cytokine production. Furthermore, IR blockade may enhance Treg cell function and survival, thereby serving as a resistance mechanism against effective immunotherapy. Given that Treg cells are comprised of functionally and phenotypically heterogeneous sub-populations that may alter their characteristics in a context-dependent manner, it is critical to identify unique molecular pathways that are preferentially used by intratumoral Treg cells. In this review, we discuss markers that serve to identify certain Treg cell subsets, distinguished by chemokine receptors, IRs and cytokines that facilitate their migration, stability and function in the TME. We also discuss how these Treg cell subsets correlate with the clinical outcome of patients with various types of cancer and how they may serve as potential TME-specific targets for novel cancer immunotherapies.
Collapse
Affiliation(s)
- Hiroshi Yano
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Graduate Program in Microbiology and Immunology (PMI), University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lawrence P Andrews
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Weingartner E, Golding A. Direct control of B cells by Tregs: An opportunity for long-term modulation of the humoral response. Cell Immunol 2017; 318:8-16. [DOI: 10.1016/j.cellimm.2017.05.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/20/2017] [Accepted: 05/28/2017] [Indexed: 12/23/2022]
|
13
|
Patient-derived xenografts of low-grade B-cell lymphomas demonstrate roles of the tumor microenvironment. Blood Adv 2017; 1:1263-1273. [PMID: 29296768 DOI: 10.1182/bloodadvances.2017005892] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/24/2017] [Indexed: 12/30/2022] Open
Abstract
To discern features of non-Hodgkin lymphomas (NHL) that are autonomous from those that are shaped by the tumor environment (TE), we used patient-derived xenografts (PDX) to probe the effects on neoplastic cells of manipulating the TE. Properties of neoplastic cells that are often considered to be autonomous include their relative independence from stromal support, their relative survival and/or proliferation advantages compared with nonneoplastic cells, and their state of differentiation. Prior approaches to creation of PDX models likely select for neoplasms, which are the most capable of engraftment, potentially masking the effects of the TE. To overcome this bias, we developed a robust protocol that rapidly produced xenografts with more than 85% of unselected, cryo-preserved, B-cell NHL specimens, including low-grade tumors such as follicular and marginal zone lymphoma. To discern features that are shaped by the TE, we extensively studied 4 low-grade lymphoma specimens. B-cell engraftment required components of the native TE; specifically, CD4+ cells. The relative survival of neoplastic compared with nonneoplastic B cells was not autonomous in 2 specimens; specifically, neoplastic B cells from 2 specimens showed a greater dependence on the TE than normal B cells for engraftment. Furthermore, the differentiation of neoplastic B cells was dependent on the TE; mature B-cell neoplasms converted to plasmacytoma-like lesions in the grafts. These results highlight the central and patient-specific roles of the TE in maintaining the relative survival of neoplastic cells compared with normal cells and in controlling the differentiation of neoplastic cells.
Collapse
|
14
|
Cai S, Hou J, Fujino M, Zhang Q, Ichimaru N, Takahara S, Araki R, Lu L, Chen JM, Zhuang J, Zhu P, Li XK. iPSC-Derived Regulatory Dendritic Cells Inhibit Allograft Rejection by Generating Alloantigen-Specific Regulatory T Cells. Stem Cell Reports 2017; 8:1174-1189. [PMID: 28434942 PMCID: PMC5425686 DOI: 10.1016/j.stemcr.2017.03.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/17/2017] [Accepted: 03/27/2017] [Indexed: 01/03/2023] Open
Abstract
Regulatory dendritic cell (DCregs)-based immunotherapy is a potential therapeutic tool for transplant rejection. We generated DCregs from murine induced pluripotent stem cells (iPSCs), which could remain in a “stable immature stage” even under strong stimulation. Harnessing this characteristic, we hypothesized that iPS-DCregs worked as a negative vaccine to generate regulatory T cells (Tregs), and induced donor-specific allograft acceptance. We immunized naive CBA (H-2Kk) mice with B6 (H-2Kb) iPS-DCregs and found that Tregs (CD4+CD25+FOXP3+) significantly increased in CBA splenocytes. Moreover, immunized CBA recipients permanently accepted B6 cardiac grafts in a donor-specific pattern. We demonstrated mechanistically that donor-type iPS-DCregs triggered transforming growth factor β1 secretion, under which the donor-antigen peptides directed naive CD4+ T cells to differentiate into donor-specific FOXP3+ Tregs instead of into effector T cells in vivo. These findings highlight the potential of iPS-DCregs as a key cell therapy resource in clinical transplantation. iPS-DCregs keep in stable immature stage that makes them a powerful cellular vaccine Donor-type iPS-DCregs lead to permanent acceptance of allogeneic cardiac grafts iPS-DCregs reduce CTL and downregulate proinflammatory cytokine iPS-DCregs enhance Tregs transmigration capability in a TGF-β1-dependent manner
Collapse
Affiliation(s)
- Songjie Cai
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Jiangang Hou
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Qi Zhang
- Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Naotsugu Ichimaru
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Shiro Takahara
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Ryoko Araki
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Lina Lu
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ji-Mei Chen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Jian Zhuang
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Ping Zhu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China.
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.
| |
Collapse
|
15
|
Xerri L, Huet S, Venstrom JM, Szafer-Glusman E, Fabiani B, Canioni D, Chassagne-Clément C, Dartigues-Cuilléres P, Charlotte F, Laurent C, Gelas-Dore B, Bolen CR, Punnoose E, Bouabdallah R, Brice P, Morschhauser F, Cartron G, Olive D, Salles G. Rituximab treatment circumvents the prognostic impact of tumor-infiltrating T-cells in follicular lymphoma patients. Hum Pathol 2017; 64:128-136. [PMID: 28414090 DOI: 10.1016/j.humpath.2017.03.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/13/2017] [Accepted: 03/26/2017] [Indexed: 01/10/2023]
Abstract
Previous immunohistochemical (IHC) studies showed controversial data about the prognostic value of tumor-infiltrating lymphocytes (TILs) in follicular lymphoma (FL). To clarify this issue, a large series of FL samples from rituximab-treated patients enrolled in the randomized PRIMA trial was examined. IHC was quantified using automated image analysis in 417, 287, 418, 406, 379, and 369 patients for CD3, CD4, CD8, PD1, ICOS, and FOXP3, respectively. RNAseq analysis was used to quantify TIL-related mRNA transcripts from 148 patients. When each IHC marker was used as a continuous variable in the whole cohort, high CD3 counts were associated with better progression-free survival (PFS) (P = .025). When an optimal IHC cut point was applied to the whole patient population, high CD3 counts and high PD1 counts were associated with better PFS (P = .011 and P = .044, respectively), whereas none of the other TIL markers had any significant correlation with outcome. When a stringent analysis was performed by dividing the whole cohort into a training set and a validation set, none of the TIL markers showed a prognostic significance in both groups. RNAseq analysis showed a significant correlation between high levels of CD3 and CD8 transcripts and better PFS (P = .001 and P = .037, respectively). No prognostic correlation was found as to the level of other immune gene transcripts. These results suggest that the IHC prognostic value of TILs is circumvented by rituximab treatment, although there is a trend for high numbers of CD3+ TILs to correlate with better PFS.
Collapse
Affiliation(s)
- Luc Xerri
- Department of Bio-Pathology, Hematology, and Tumor Immunology, Institut Paoli-Calmettes and Aix-Marseille Univ, Marseille, France; Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, Marseille, F-13009, France; CNRS, UMR7258, Marseille, F-13009, Institut Paoli-Calmettes, Marseille, F-13009, France; Aix-Marseille University, UM, 105, F-13284, Marseille, France.
| | - Sarah Huet
- INSERM1052, CNRS 5286, Centre de Recherche en Cancerologie de Lyon, Faculté de Médecine Lyon-Sud Charles Mérieux, Hospices Civils De Lyon, Laboratoire d'hématologie, F-69495 Pierre Bénite cedex
| | | | | | - Bettina Fabiani
- Department of Pathology, Centre Hospitalier Saint Antoine, F-75571 Paris, France
| | - Danielle Canioni
- Department of Pathology, Centre Hospitalier Necker, F-75743 Paris, France
| | | | | | - Fréderic Charlotte
- Department of Pathology, Centre Hospitalier Pitié-Salpêtriére, F-75651 France
| | - Camille Laurent
- Department of Pathology, INSERM U.1037, Institut Universitaire du Cancer-Oncopole, F-31059 Toulouse
| | - Benedicte Gelas-Dore
- INSERM1052, CNRS 5286, Centre de Recherche en Cancerologie de Lyon, Faculté de Médecine Lyon-Sud Charles Mérieux, Hospices Civils De Lyon, Laboratoire d'hématologie, F-69495 Pierre Bénite cedex
| | | | | | - Reda Bouabdallah
- Department of Bio-Pathology, Hematology, and Tumor Immunology, Institut Paoli-Calmettes and Aix-Marseille Univ, Marseille, France; Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, Marseille, F-13009, France; CNRS, UMR7258, Marseille, F-13009, Institut Paoli-Calmettes, Marseille, F-13009, France; Aix-Marseille University, UM, 105, F-13284, Marseille, France
| | - Pauline Brice
- Department of Hematology, Centre Hospitalier Saint Louis, F-75475 Paris, France
| | - Franck Morschhauser
- Department of Hematology, Hopital Claude Huriez, Unité GRITA, Université de Lille 2, F-59000 Lille, France
| | - Guillaume Cartron
- Departement of Hematology, CHU Montpellier, UMR CNRS-5235, F-34295 Montpellier, France
| | - Daniel Olive
- Department of Bio-Pathology, Hematology, and Tumor Immunology, Institut Paoli-Calmettes and Aix-Marseille Univ, Marseille, France; Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, Marseille, F-13009, France; CNRS, UMR7258, Marseille, F-13009, Institut Paoli-Calmettes, Marseille, F-13009, France; Aix-Marseille University, UM, 105, F-13284, Marseille, France
| | - Gilles Salles
- INSERM1052, CNRS 5286, Centre de Recherche en Cancerologie de Lyon, Faculté de Médecine Lyon-Sud Charles Mérieux, Hospices Civils De Lyon, Laboratoire d'hématologie, F-69495 Pierre Bénite cedex
| | | |
Collapse
|
16
|
Checkpoint Inhibition: Programmed Cell Death 1 and Programmed Cell Death 1 Ligand Inhibitors in Hodgkin Lymphoma. Cancer J 2016; 22:17-22. [PMID: 26841012 DOI: 10.1097/ppo.0000000000000164] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Hodgkin lymphoma (HL) is a lymphoid malignancy characterized by a reactive immune infiltrate surrounding relatively few malignant cells. In this scenario, active immune evasion seems to play a central role in allowing tumor progression. Immune checkpoint inhibitor pathways are normal mechanisms of T-cell regulation that suppress immune effector function following an antigenic challenge. Hodgkin lymphoma cells are able to escape immune surveillance by co-opting these mechanisms. The programmed cell death 1 (PD-1) pathway in particular is exploited in HL as the malignant Hodgkin and Reed-Sternberg cells express on their surface cognate ligands (PD-L1/L2) for the PD-1 receptor and thereby dampen the T-cell-mediated antitumoral response. Monoclonal antibodies that interact with and disrupt the PD-1:PD-L1/L2 axis have now been developed and tested in early-phase clinical trials in patients with advanced HL with encouraging results. The remarkable clinical activity of PD-1 inhibitors in HL highlights the importance of immune checkpoint pathways as therapeutic targets in HL. In this review, we discuss the rationale for targeting PD-1 and PD-L1 in the treatment of HL. We will evaluate the published clinical data on the different agents and highlight the safety profile of this class of agents. We discuss the available evidence on the use of biomarkers as predictors of response to checkpoint blockade and summarize the areas under active investigation in the use of PD-1/PD-L1 inhibitors for the treatment of HL.
Collapse
|
17
|
Ansell SM. Where Do Programmed Death-1 Inhibitors Fit in the Management of Malignant Lymphoma? J Oncol Pract 2016; 12:101-6. [PMID: 26869644 DOI: 10.1200/jop.2015.009191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tumor-specific cytotoxic T cells have the capacity to target and eradicate malignant B cells in patients with Hodgkin and non-Hodgkin lymphoma; however, multiple mechanisms, including regulatory T cells, immunosuppressive ligands, and immune exhaustion, suppress an effective antitumor immune response. One mechanism that is used by malignant cells to inhibit the immune response is overexpression of programmed death ligand 1 or 2 (PD-L1 or PD-L2) on the cancer cell surface. These ligands interact with the programmed death-1 (PD-1) receptor expressed on intratumoral T cells and provide an inhibitory signal, thereby suppressing the antitumor immune response. Monoclonal antibodies that block PD-1 signaling prevent T-cell inhibition and promote a T-cell-mediated antilymphoma response. Blocking antibodies that are directed against PD-1 or PD-L1 are currently being tested in patients with lymphoma and have shown remarkable efficacy, particularly in patients with relapsed Hodgkin lymphoma. On the basis of the promising activity of this approach, PD-1 inhibitors are being used as single-agent therapy in patients with relapsed Hodgkin lymphoma, and these inhibitors are also being tested in combination with standard chemotherapy or targeted agents in ongoing clinical trials.
Collapse
|
18
|
Abstract
The immune system has evolved to defend the organism against an almost infinite number of pathogens in a locally confined and antigen-specific manner while at the same time preserving tolerance to harmless antigens and self. Regulatory T (Treg) cells essentially contribute to an immunoregulatory network preventing excessive immune responses and immunopathology. There is emerging evidence that Treg cells not only operate in secondary lymphoid tissue but also regulate immune responses directly at the site of inflammation. Hence, the classification of Treg cells might need to be further extended by Treg cell subsets that are functionally and phenotypically polarized by their residency. In this review, we discuss recent findings on these tissue-resident Treg cell subsets and how these cells may operate in a tissue- and context-dependent manner.
Collapse
|
19
|
Le KS, Thibult ML, Just-Landi S, Pastor S, Gondois-Rey F, Granjeaud S, Broussais F, Bouabdallah R, Colisson R, Caux C, Ménétrier-Caux C, Leroux D, Xerri L, Olive D. Follicular B Lymphomas Generate Regulatory T Cells via the ICOS/ICOSL Pathway and Are Susceptible to Treatment by Anti-ICOS/ICOSL Therapy. Cancer Res 2016; 76:4648-60. [PMID: 27246829 DOI: 10.1158/0008-5472.can-15-0589] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/09/2016] [Indexed: 11/16/2022]
Abstract
The prognosis of follicular lymphoma (FL) patients is suspected to be influenced by tumor-infiltrating regulatory T cells (Treg). The mechanism of Treg enrichment in FL and their impact on malignant FL B cells remains to be elucidated. We analyzed 46 fresh lymph node biopsy samples, including FL (n = 20), diffuse large B-cell lymphoma (n = 10), classical Hodgkin lymphoma (n = 9), and reactive lymphadenitis (n = 7). Using multicolor flow cytometry and cell sorting, we observed an accumulation of CD25(high)CD127(low/neg) Tregs in FL tissues. These Tregs comprised activated ICOS(+) Tregs that were able to suppress not only conventional T cells, but also FL B cells. These FL B cells were able to express ICOSL in vitro and to generate CD25(high)FoxP3(high) Tregs expressing ICOS. Treg generation was associated with ICOS/ICOSL engagement and was abrogated by antagonist anti-ICOS and anti-ICOSL antibodies. Interactions between Tregs and FL B cells resulted in ICOSL downregulation on FL B cells. Our results highlight a key role for Tregs in FL pathogenesis and suggest that targeting the ICOS/ICOSL pathway may be a promising immunotherapy for FL treatment. Cancer Res; 76(16); 4648-60. ©2016 AACR.
Collapse
Affiliation(s)
- Kieu-Suong Le
- Centre de recherche en Cancérologie de Marseille, Inserm U1068/CNRS U7258, Marseille, France. Aix Marseille Université, Marseille, France
| | - Marie-Laure Thibult
- Centre de recherche en Cancérologie de Marseille, Inserm U1068/CNRS U7258, Marseille, France. Institut Paoli - Calmettes, Marseille, France
| | | | - Sonia Pastor
- Centre de recherche en Cancérologie de Marseille, Inserm U1068/CNRS U7258, Marseille, France
| | - Françoise Gondois-Rey
- Centre de recherche en Cancérologie de Marseille, Inserm U1068/CNRS U7258, Marseille, France
| | - Samuel Granjeaud
- Centre de recherche en Cancérologie de Marseille, Inserm U1068/CNRS U7258, Marseille, France
| | | | | | - Renaud Colisson
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052/CNRS 5286, Lyon, France
| | - Christophe Caux
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052/CNRS 5286, Lyon, France
| | | | | | - Luc Xerri
- Centre de recherche en Cancérologie de Marseille, Inserm U1068/CNRS U7258, Marseille, France. Aix Marseille Université, Marseille, France. Institut Paoli - Calmettes, Marseille, France
| | - Daniel Olive
- Centre de recherche en Cancérologie de Marseille, Inserm U1068/CNRS U7258, Marseille, France. Aix Marseille Université, Marseille, France. Institut Paoli - Calmettes, Marseille, France.
| |
Collapse
|
20
|
Nedelkovska H, Rosenberg AF, Hilchey SP, Hyrien O, Burack WR, Quataert SA, Baker CM, Azadniv M, Welle SL, Ansell SM, Kim M, Bernstein SH. Follicular Lymphoma Tregs Have a Distinct Transcription Profile Impacting Their Migration and Retention in the Malignant Lymph Node. PLoS One 2016; 11:e0155347. [PMID: 27228053 PMCID: PMC4882026 DOI: 10.1371/journal.pone.0155347] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 04/06/2016] [Indexed: 01/25/2023] Open
Abstract
We have previously shown that regulatory T cells (Tregs) infiltrating follicular lymphoma lymph nodes are quantitatively and qualitatively different than those infiltrating normal and reactive nodes. To gain insight into how such Treg populations differ, we performed RNA sequence (RNAseq) analyses on flow sorted Tregs from all three sources. We identify several molecules that could contribute to the observed increased suppressive capacity of follicular lymphoma nodal tregs, including upregulation of CTLA-4, IL-10, and GITR, all confirmed by protein expression. In addition, we identify, and confirm functionally, a novel mechanism by which Tregs target to and accumulate within a human tumor microenvironment, through the down regulation of S1PR1, SELL (L-selectin) and CCR7, potentially resulting in greater lymph node retention. In addition we identify and confirm functionally the upregulation of the chemokine receptor CXCR5 as well as the secretion of the chemokines CXCL13 and IL-16 demonstrating the unique ability of the follicular derived Tregs to localize and accumulate within not only the malignant lymph node, but also localize and accumulate within the malignant B cell follicle itself. Such findings offer significant new insights into how follicular lymphoma nodal Tregs may contribute to the biology of follicular lymphoma and identify several novel therapeutic targets.
Collapse
Affiliation(s)
- Hristina Nedelkovska
- James P. Wilmot Cancer Center, Lymphoma Biology Program, Department of Medicine University of Rochester Medical Center, Rochester, New York, United States of America
| | - Alexander F. Rosenberg
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Shannon P. Hilchey
- James P. Wilmot Cancer Center, Lymphoma Biology Program, Department of Medicine University of Rochester Medical Center, Rochester, New York, United States of America
| | - Ollivier Hyrien
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - W. Richard Burack
- James P. Wilmot Cancer Center, Lymphoma Biology Program, Department of Medicine University of Rochester Medical Center, Rochester, New York, United States of America
| | - Sally A. Quataert
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Christina M. Baker
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Mitra Azadniv
- James P. Wilmot Cancer Center, Lymphoma Biology Program, Department of Medicine University of Rochester Medical Center, Rochester, New York, United States of America
| | - Stephen L. Welle
- University of Rochester Genomics Research Center, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Stephen M. Ansell
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Minsoo Kim
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester New York, United States of America
| | - Steven H. Bernstein
- James P. Wilmot Cancer Center, Lymphoma Biology Program, Department of Medicine University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW The purpose of this article is to discuss the tumor microenvironment in lymphoma, and to review potential immune targets that are now becoming relevant because of clinical responses seen with the use of immune checkpoint inhibitors. RECENT FINDINGS Recent data have shown that cells within the immune microenvironment in lymphoma express programmed death ligand-1 (PD-L1) and many of the intratumoral T cells with an exhausted immune phenotype express programmed cell death-1 (PD-1). This provides a novel opportunity to inhibit the immune checkpoints and initial clinical trials utilizing antibodies that block the interaction between PD-1 and PD-L1 have demonstrated significant clinical responses in various lymphomas, including Hodgkin lymphoma. SUMMARY The use of immune checkpoint inhibitors, including nivolumab and pembrolizumab, in relapsed and refractory patients with lymphoma is proving highly successful. Patients with Hodgkin lymphoma, in particular, have a very high response rate to PD-1 blockade and responses in these patients appear durable.
Collapse
|
22
|
Villasboas JC, Ansell SM. Nivolumab for the treatment of classical Hodgkin lymphoma after failure of autologous stem cell transplant and brentuximab. Expert Rev Anticancer Ther 2015; 16:5-12. [PMID: 26577822 DOI: 10.1586/14737140.2016.1121812] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cancer cells are able to escape surveillance from the immune system by co-opting physiologic mechanisms such as the programmed cell death-1 (PD-1) receptor pathway. Agents able to block the interaction between the PD-1 receptor and its ligands have the potential to release T cells from tumor-induced suppression and eradicate malignant cells. Nivolumab - a PD-1 inhibitor - is approved for the treatment of patients with metastatic melanoma and lung cancer. This agent has been tested in patients with advanced Hodgkin lymphoma (HL) and showed impressive results in a phase I trial. Here we review the profile of Nivolumab including its pharmacological properties, clinical efficacy and safety in patients with advanced classical HL.
Collapse
Affiliation(s)
- Jose C Villasboas
- a Department of Medicine/Division of Hematology , Mayo Clinic , Rochester , MN , USA
| | - Stephen M Ansell
- a Department of Medicine/Division of Hematology , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
23
|
Yang ZZ, Liang AB, Ansell SM. T-cell-mediated antitumor immunity in B-cell non-Hodgkin lymphoma: activation, suppression and exhaustion. Leuk Lymphoma 2015; 56:2498-504. [PMID: 25651421 DOI: 10.3109/10428194.2015.1011640] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The tumor microenvironment in B-cell non-Hodgkin lymphoma (NHL) comprises not only malignant cells but also significant numbers of normal immune cells. The intratumoral immune infiltrate includes T-lymphocytes that appear to target the malignant clone. Despite immunologically recognizing the lymphoma cells, the intratumoral T-cells are unable to eradicate the malignant cells and the lymphoma commonly progresses. Recent data has identified mechanisms whereby activated intratumoral T-cells are suppressed or become exhausted due to chronic antigen stimulation. A clearer understanding of these mechanisms will allow for strategies to overcome them and improve the outcome of patients with lymphoma.
Collapse
Affiliation(s)
- Zhi-Zhang Yang
- a Division of Hematology and Internal Medicine, Mayo Clinic , Rochester , MN , USA
| | - Ai-Bin Liang
- b Department of Hematology , Tongji Hospital, Tongji University , Shanghai , China
| | - Stephen M Ansell
- a Division of Hematology and Internal Medicine, Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
24
|
PD-1 expression defines two distinct T-cell sub-populations in follicular lymphoma that differentially impact patient survival. Blood Cancer J 2015; 5:e281. [PMID: 25700246 PMCID: PMC4349259 DOI: 10.1038/bcj.2015.1] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 12/30/2014] [Indexed: 01/27/2023] Open
Abstract
To determine the biological and clinical relevance of programmed death 1 (PD-1) in follicular lymphoma (FL), we characterized PD-1+ T-cell subsets and assessed their biological function as well as potential clinical impact. We found that PD-1 is expressed on intratumoral CD4+ T cells with both bright and dim intensity, representing two different sub-populations of cells. By immunohistochemistry, we found that CD4+PD-1high T cells predominantly reside in the lymph node follicles, while PD-1low T cells are mainly located in an interfollicular pattern. Intratumoral CD4+PD-1high T cells have a TFH cell phenotype, express CXCR5, secrete IL-21 and are BCL-6 positive with no TIM-3 expression. In contrast, CD4+PD-1low T cells have an exhausted phenotype, express TIM-3 and do not express BCL-6 and CXCR5. Functionally, CD4+PD-1high T cells actively supported B-cell growth, while CD4+PD-1low T cells displayed a reduced cytokine production and cell-signal transduction. Clinically, we observed that the numbers of CD4+ or CD8+PD-1low T cells significantly correlate with a reduced overall survival in FL patients (P=0.007 and 0.04 respectively; n=32). In contrast, the number of CD4+PD-1high T cells was not associated with patient outcome. Taken together, these results indicated that PD-1 expression defines two sub-populations with distinct functions that differentially impact patient outcome in FL.
Collapse
|
25
|
Brady MT, Hilchey SP, Hyrien O, Spence SA, Bernstein SH. Mesenchymal stromal cells support the viability and differentiation of follicular lymphoma-infiltrating follicular helper T-cells. PLoS One 2014; 9:e97597. [PMID: 24836297 PMCID: PMC4023957 DOI: 10.1371/journal.pone.0097597] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 04/21/2014] [Indexed: 12/20/2022] Open
Abstract
The biology of follicular lymphoma (FL) is largely dictated by the immune-effector and stromal cells that comprise its tumor microenvironment. FL-infiltrating T-cell populations that are thought to be fundamental to FL biology are follicular helper T-cells (TFH), follicular regulatory T-cells (TFR), a recently described population that regulates TFH activity, and regulatory T-cells (Treg). These T-cell populations have dynamic interactions with mesenchymal stromal cells (MSCs) in the tumor microenvironment. Whereas MSCs have been shown to support FL B-cell and Treg viability, their effects on FL-infiltrating TFH and TFR cells have not been described. Herein we show that MSCs support the viability of FL-infiltrating TFH and TFR, as well as Tregs, in part through an IL-6-dependent mechanism. We further demonstrate that MSCs mediate TFH to TFR conversion by inducing the expression of FoxP3 in TFH cells, demonstrating for the first time that human TFR can be derived from TFH cells. Given that the balance of TFH and TFR populations likely dictate, in part, the biology of this disease, our data support the potential for targeting MSCs as a therapeutic strategy.
Collapse
Affiliation(s)
- Michael T. Brady
- James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Shannon P. Hilchey
- James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Ollivier Hyrien
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Stephen A. Spence
- James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Steven H. Bernstein
- James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
26
|
Voo KS, Foglietta M, Percivalle E, Chu F, Nattamai D, Harline M, Lee ST, Bover L, Lin HY, Baladandayuthapani V, Delgado D, Luong A, Davis RE, Kwak LW, Liu YJ, Neelapu SS. Selective targeting of Toll-like receptors and OX40 inhibit regulatory T-cell function in follicular lymphoma. Int J Cancer 2014; 135:2834-46. [PMID: 24771328 DOI: 10.1002/ijc.28937] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 03/25/2014] [Indexed: 01/08/2023]
Abstract
Immunotherapeutic strategies are promising approaches for the treatment of follicular lymphoma (FL). However, their efficacy may be limited by immunosuppressive elements in the immune system and tumor microenvironment. Therefore, strategies to reverse the effects of the immunosuppressive elements are needed. We observed that regulatory T cells (Tregs) were increased in the peripheral blood at diagnosis and persisted in high numbers after induction of clinical remission with a cyclophosphamide and doxorubicin-containing chemotherapy regimen in FL patients. High levels of peripheral blood Tregs prior to therapy were associated with decreased progression-free survival in FL patients treated with either chemotherapy or combination immunotherapy that targeted CD20 and PD-1 with monoclonal antibodies rituximab and pidilizumab, respectively. Intratumoral and peripheral blood Tregs potently suppressed autologous antitumor effector T cells in FL. However, the effects of FL Tregs could be reversed by triggering Toll-like receptors (TLR) with TLR ligands Pam3 CSK4 (TLR 1/2), flagellin (TLR 5), and CpG-B (TLR 9), and/or OX40. The TLR ligands synergized with each other as well as OX40 signaling to inhibit Tregs. Furthermore, they restored the function of FL tumor-specific effector T cells. Our results suggest that a state of tolerance exists in FL patients at diagnosis and after induction of clinical remission, and agents that activate TLRs 1/2, 5, and 9, and OX40 may serve as adjuvants to enhance the efficacy of antitumor immunotherapeutic strategies and preventive vaccines against infectious diseases in these patients.
Collapse
Affiliation(s)
- Kui Shin Voo
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX; Center for Cancer Immunology Research, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Procházka V, Papajík T, Faber E, Raida L, Kapitáňová Z, Langová K, Prouzová Z, Jarošová M, Indrák K. Soluble interleukin-2 receptor level predicts survival in patients with follicular lymphoma treated with cyclophosphamide, doxorubicin, vincristine and prednisone chemotherapy in the rituximab era. Leuk Lymphoma 2014; 55:1584-90. [DOI: 10.3109/10428194.2013.850167] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
28
|
Sakai A, Yoshida N. The Role of Tumor-Associated Macrophages on Serum Soluble IL-2R Levels in B-Cell Lymphomas. J Clin Exp Hematop 2014; 54:49-57. [DOI: 10.3960/jslrt.54.49] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
29
|
Hua F, Kang N, Gao YA, Cui LX, Ba DN, He W. Potential regulatory role of in vitro-expanded Vδ1 T cells from human peripheral blood. Immunol Res 2013; 56:172-80. [PMID: 23532670 DOI: 10.1007/s12026-013-8390-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
γδ T cells represent a relevant proportion of T lymphocytes that express T cell receptors (TCRs) encoded by the γ and δ T cell receptor genes. Whereas the most circulating γδ T cell type, Vδ2 T cells, has been described and studied intensively, the potential role of Vδ1 T cells remains largely unclear. Here, we identified that expanded peripheral blood Vδ1 T cells stimulated with anti-human TCR Vδ1 monoclonal antibody (mAb) in vitro predominantly expressed forkhead box p3 (Foxp3). In addition, these cells also expressed other regulatory T cell-related molecules, such as CD25, glucocorticoid-induced TNFR family-related protein and cytotoxic T lymphocyte-associated protein-4 (CTLA-4), and held the potent capacity for the production of transforming growth factor beta 1 (TGF-β1). These autocrine and/or paracrine TGF-β1 could bind TGF-β1 receptors on Vδ1 T cells and induce sustained Foxp3 expression. Moreover, Foxp3 expression coincided with high CD25 expression. CD25(high) Vδ1 T cells exhibited stronger suppression on CD4(+) T cell proliferation compared with TGF-β1-induced CD25(high) CD4(+) regulatory T cells. Therefore, our phenotypic and functional analyses first demonstrate the potential regulatory property of anti-human TCR Vδ1 mAb-activated Vδ1 T cells. These results will broaden our understanding about the role of peripheral blood Vδ1 T cells under physical and pathological conditions.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Blood Cells/immunology
- Blood Circulation
- CTLA-4 Antigen/metabolism
- Cell Proliferation
- Cells, Cultured
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Humans
- Immunosuppression Therapy
- Interleukin-2 Receptor alpha Subunit/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- T-Lymphocytes, Regulatory/immunology
- Transforming Growth Factor beta1/immunology
- Transforming Growth Factor beta1/metabolism
- Young Adult
Collapse
Affiliation(s)
- Fang Hua
- Department of Immunology, School of Basic Medicine, Peking Union Medical College, National Key Laboratory of Medical Molecular Biology, Chinese Academy of Medical Sciences, Beijing, China.
| | | | | | | | | | | |
Collapse
|
30
|
Zheng S, Shen Y, Song Y, Yuan Y. How to detour Treg cells in T cell-based antitumor immune therapy. Onco Targets Ther 2013; 6:1243-7. [PMID: 24043948 PMCID: PMC3772754 DOI: 10.2147/ott.s48872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
T cell-based antitumor immune therapy which occupies the boosting area of translational medicine research is capable of eradicating some kinds of tumors that are in late stages. However, the effectiveness of adoptive cell transfer treatment varies among the different clinical trials, while the safety of cells is still uncertain for some patients. All these phenomena provoke us to ask whether the instability of T cell-based antitumor immune therapy is due to immune modulation function of Treg cells in the tumor microenvironment and the peripheral circulation. Some successful Treg-targeting treatments in clinical trials provide the inspiration for subtle modulation of Treg cells in future cancer immunotherapies. We hypothesized that Treg cells may somehow sense the abundance of peripheral immune effector cells, and maintain the shifted tumor-bearing homeostasis of the immune system. Killer cells infused in adoptive cell transfer therapy may be monitored and spontaneously downregulated by Treg cells. Further studies are required to develop more effective combinations of immunotherapy with conventional chemo/radiotherapy in the modulation of immune-suppressive cells.
Collapse
Affiliation(s)
- Shu Zheng
- The Cancer Institute, Key Laboratory of Cancer Prevention and Intervention China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | | | | | | |
Collapse
|
31
|
Burocchi A, Colombo MP, Piconese S. Convergences and divergences of thymus- and peripherally derived regulatory T cells in cancer. Front Immunol 2013; 4:247. [PMID: 23986759 PMCID: PMC3753661 DOI: 10.3389/fimmu.2013.00247] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/08/2013] [Indexed: 12/18/2022] Open
Abstract
The expansion of regulatory T cells (Treg) is a common event characterizing the vast majority of human and experimental tumors and it is now well established that Treg represent a crucial hurdle for a successful immunotherapy. Treg are currently classified, according to their origin, into thymus-derived Treg (tTreg) or peripherally induced Treg (pTreg) cells. Controversy exists over the prevalent mechanism accounting for Treg expansion in tumors, since both tTreg proliferation and de novo pTreg differentiation may occur. Since tTreg and pTreg are believed as preferentially self-specific or broadly directed to non-self and tumor-specific antigens, respectively, the balance between tTreg and pTreg accumulation may impact on the repertoire of antigen specificities recognized by Treg in tumors. The prevalence of tTreg or pTreg may also affect the outcome of immunotherapies based on tumor-antigen vaccination or Treg depletion. The mechanisms dictating pTreg induction or tTreg expansion/stability are a matter of intense investigation and the most recent results depict a complex landscape. Indeed, selected Treg subsets may display peculiar characteristics in terms of stability, suppressive function, and cytokine production, depending on microenvironmental signals. These features may be differentially distributed between pTreg and tTreg and may significantly affect the possibility of manipulating Treg in cancer therapy. We propose here that innovative immunotherapeutic strategies may be directed at diverting unstable/uncommitted Treg, mostly enriched in the pTreg pool, into tumor-specific effectors, while preserving systemic immune tolerance ensured by self-specific tTreg.
Collapse
Affiliation(s)
- Alessia Burocchi
- Molecular Immunology Unit, Department of Experimental Medicine, Fondazione IRCCS "Istituto Nazionale Tumori," Milan , Italy
| | | | | |
Collapse
|
32
|
Van Herwijnen MJC, Van Der Zee R, Van Eden W, Broere F. Heat shock proteins can be targets of regulatory T cells for therapeutic intervention in rheumatoid arthritis. Int J Hyperthermia 2013; 29:448-54. [PMID: 23863094 DOI: 10.3109/02656736.2013.811546] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterised by excessive immune responses resulting in inflammation of the joints. Although current therapies can be successful in dampening inflammation, a long-lived state of tolerance is seldom achieved. Therefore, novel therapies are needed that restore and maintain tolerance in patients with RA. Targeting regulatory T cells (Tregs) is a successful strategy to achieve tolerance, as was shown in studies performed in animal models and in human clinical trials. The antigen-specificity of Tregs is crucial for their effectiveness and allows for very specific targeting of these cells. However, which antigen is suitable for autoimmune diseases such as RA, for which the autoantigens are largely unknown? Heat shock proteins (HSPs) are ubiquitously expressed and can be up-regulated during inflammation. Additionally, HSPs, or HSP-derived peptides are immunogenic and can be recognised by a variety of immune cells, including Tregs. Therefore, this review highlights the potential of HSP-specific Tregs to control inflammatory immune responses. Targeting HSP-specific Tregs in RA can be achieved via the administration of HSPs (derived peptides), thereby controlling inflammatory responses. This makes HSPs attractive candidates for therapeutic intervention in chronic autoimmune diseases, with the ultimate goal of inducing long-lasting tolerance.
Collapse
|
33
|
Rawal S, Chu F, Zhang M, Park HJ, Nattamai D, Kannan S, Sharma R, Delgado D, Chou T, Lin HY, Baladandayuthapani V, Luong A, Vega F, Fowler N, Dong C, Davis RE, Neelapu SS. Cross talk between follicular Th cells and tumor cells in human follicular lymphoma promotes immune evasion in the tumor microenvironment. THE JOURNAL OF IMMUNOLOGY 2013; 190:6681-93. [PMID: 23686488 DOI: 10.4049/jimmunol.1201363] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The microenvironment of human follicular lymphoma (FL), an incurable B cell non-Hodgkin's lymphoma, is thought to play a major role in its pathogenesis and course. Microenvironmental cells of likely importance include follicular Th cells (TFH) and regulatory T cells (Tregs), and understanding their interactions with FL tumor cells is necessary to develop novel therapeutic strategies. We found that IL-4 and CD40L are expressed by intratumoral TFH and induce production of CCL17 and CCL22 by FL tumor cells. IL-4 alone induces only CCL17 but enhances stimulation by CD40L of both CCL17 and CCL22. Consistent with our in vitro results, mRNA transcripts of IL-4 correlated with CCL17, but not CCL22, in gene expression profiling studies of FL biopsies, whereas CD40L correlated with both CCL17 and CCL22. Tumor supernatants induced preferential migration of Tregs and IL-4-producing T cells rather than IFN-γ-producing T cells, and Abs to CCR4 significantly abrogated the migration of Tregs. Our results suggest that through two distinct mechanisms, intratumoral TFH induce production of CCL17 and CCL22 by FL tumor cells and facilitate active recruitment of Tregs and IL-4-producing T cells, which, in turn, may stimulate more chemokine production in a feed-forward cycle. Thus, TFH appear to play a major role in generating an immunosuppressive tumor microenvironment that promotes immune escape and tumor survival and growth. Our results provide novel insights into the cross talk among TFH, tumor cells, and Tregs in FL, and offer potential targets for development of therapeutic strategies to overcome immune evasion.
Collapse
Affiliation(s)
- Seema Rawal
- Division of Cancer Medicine, Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hamilton A, Sibson NR. Role of the systemic immune system in brain metastasis. Mol Cell Neurosci 2013; 53:42-51. [PMID: 23073146 DOI: 10.1016/j.mcn.2012.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 09/24/2012] [Accepted: 10/04/2012] [Indexed: 10/27/2022] Open
Abstract
Metastatic disease in the central nervous system (CNS) is a cause of increasing mortality amongst cancer patients. As with other types of cancer, cells of the systemic immune system play a range of important roles in the development of metastatic lesions in the CNS, both repressing and promoting tumour growth. Recent advances in immunotherapy have changed the emphasis in cancer treatment away from conventional chemotherapy and radiotherapy for certain tumour types. Despite this, our understanding of systemic immune system involvement in CNS metastases remains poor. The blood-brain barrier prevents the majority of diagnostic and therapeutic agents from crossing into the brain parenchyma until the late stages of metastatic disease. Thus, the development of immunotherapy for CNS pathologies is particularly desirable. This review draws together our current understanding in the relationships between CNS metastases and circulating systemic immune cells. We discuss the roles that circulating systemic immune cells may play in the homing of metastatic cells to the perivascular space, and the pro-metastatic and antagonistic roles that infiltrating systemic immune cells may play at sites of metastasis. This article is part of a Special Issue entitled 'Neuroinflammation in neurodegeneration and neurodysfunction'.
Collapse
Affiliation(s)
- Alastair Hamilton
- CR-UK/MRC Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, UK
| | | |
Collapse
|
35
|
Abstract
In addition to malignant cells, the tumor microenvironment also includes nonmalignant cells, secreted proteins, and blood vessels that surround and support the growth of the tumor. Interactions between the various components of the tumor microenvironment are significant; tumor cells can change the nature of the microenvironment, and conversely, the microenvironment can affect how a tumor grows and spreads. The structure and composition of the tumor microenvironment varies among different types of cancers and between patients. This paper focuses on the composition and function of the tumor microenvironment in hematologic malignancies with a specific focus on B-cell lymphomas.
Collapse
Affiliation(s)
- Stephen M Ansell
- From the Division of Hematology, Mayo Clinic, Rochester, MN; Abrahamson Cancer Center of the University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
36
|
Podnos A, Clark DA, Erin N, Yu K, Gorczynski RM. Further evidence for a role of tumor CD200 expression in breast cancer metastasis: decreased metastasis in CD200R1KO mice or using CD200-silenced EMT6. Breast Cancer Res Treat 2012; 136:117-27. [PMID: 23053647 DOI: 10.1007/s10549-012-2258-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 09/12/2012] [Indexed: 11/27/2022]
Abstract
Previous studies reported that CD200 expression on cells of the transplantable EMT6 mouse breast cancer line was increased during growth in immunocompetent mice. Low levels of expression persisted in NOD-SCID.IL-2(γr-/-) mice or mice with generalized over-expression of a CD200 transgene (CD200(tg) mice), despite the faster tumor growth in both of these latter strains. We also showed that CD200 expression (by the host and/or tumor cells) led to increased seeding of tumor cells to DLN in immunocompromised (CD200(tg) or NOD-SCID.IL-2(γr-/-)) vs immunocompetent mice, using limiting dilution cloning of tumor cells from DLN (vs contralateral lymph nodes, CLN). Evidence for an important role for CD200 expression in this increased metastasis came from the observation that neutralization of CD200 by anti-CD200mAbs decreased tumor metastasis and increased levels of cytotoxic anti-tumor immune cells in DLN. In the current studies, we have extended these observations by exploring tumor growth/metastasis in CD200R1 KO mice in which we have previously shown, in a transplant model, that expression of CD200 fails to deliver an immunosuppressive signal. In addition, we have studied local and metastatic growth in healthy control mice of EMT6 tumor cells stably transduced with shRNA able to silence CD200 expression. In both scenarios, decreased metastasis was observed, with increased immunity to EMT6 detected by cytotoxicity assays. In addition, adoptive transfer of DLN to control mice attenuated EMT6 metastases implying a potential therapeutic benefit from neutralizing CD200 expression in breast cancer.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Breast Neoplasms/immunology
- Breast Neoplasms/therapy
- Female
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immunotherapy
- Interleukin-2/immunology
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/immunology
- Mammary Neoplasms, Animal/therapy
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Neoplasm Metastasis/immunology
- Neoplasm Metastasis/pathology
- Neoplasm Metastasis/therapy
Collapse
Affiliation(s)
- Anna Podnos
- University Health Network, Toronto General Hospital, 101 College Street, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
37
|
Yang ZZ, Grote DM, Ziesmer SC, Niki T, Hirashima M, Novak AJ, Witzig TE, Ansell SM. IL-12 upregulates TIM-3 expression and induces T cell exhaustion in patients with follicular B cell non-Hodgkin lymphoma. J Clin Invest 2012; 122:1271-82. [PMID: 22426209 DOI: 10.1172/jci59806] [Citation(s) in RCA: 218] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 02/01/2012] [Indexed: 12/18/2022] Open
Abstract
The cytokine IL-12 induces IFN-γ production by T and NK cells. In preclinical models, it contributes to antitumor immunity. However, in clinical testing, it has shown limited benefit in patients with any one of a variety of malignancies. Moreover, in a clinical trial testing a combination of IL-12 and rituximab in patients with follicular B cell non-Hodgkin lymphoma (FL), those treated with IL-12 showed a lower response rate, suggesting that IL-12 actually plays a detrimental role. Here, we investigated whether the failure of IL-12 treatment for FL was due to T cell exhaustion, a condition characterized by reduced T cell differentiation, proliferation, and function, which has been observed in chronic viral infection. We found that extended exposure to IL-12 induced T cell exhaustion and contributed to the poor prognosis in FL patients. Long-term exposure of freshly isolated human CD4+ T cells to IL-12 in vitro caused T cell dysfunction and induced expression of TIM-3, a T cell immunoglobulin and mucin domain protein with a known role in T cell exhaustion, via an IFN-γ-independent mechanism. TIM-3 was required for the negative effect of IL-12 on T cell function. Importantly, TIM-3 also was highly expressed on intratumoral T cells that displayed marked functional impairment. Our findings identify IL-12- and TIM-3-mediated exhaustion of T cells as a mechanism for poor clinical outcome when IL-12 is administered to FL patients.
Collapse
Affiliation(s)
- Zhi-Zhang Yang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Entourage: the immune microenvironment following follicular lymphoma. Blood Cancer J 2012; 2:e52. [PMID: 22829236 PMCID: PMC3270257 DOI: 10.1038/bcj.2011.53] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 11/30/2011] [Accepted: 12/08/2011] [Indexed: 12/17/2022] Open
Abstract
In follicular lymphoma, nonmalignant immune cells are important. Follicular lymphoma depends on CD4+ cells, but CD8+ cells counteract it. We hypothesized that the presence of follicular lymphoma is associated with higher CD4+ than CD8+ cell numbers in the tumor microenvironment but not in the immune system. Using flow cytometry, pre-treatment and follow-up CD4/CD8 ratios were estimated in the bone marrow, blood and lymph nodes of untreated follicular lymphoma patients in two independent data sets (N1=121; N2=166). The ratios were analyzed for their relation with bone marrow lymphoma involvement. Bone marrows were also investigated with immunohistochemistry. In either data set, the bone marrow CD4/CD8 ratios were higher in bone marrows involved with lymphoma (P=0.043 and 0.0002, respectively). The mean CD4/CD8 ratio was 1.0 in uninvolved and 1.4 in involved bone marrows. Also higher in involved bone marrows were CD4/CD56 and CD3CD25/CD3 ratios. No blood or lymph node ratios differed between bone marrow-negative and -positive patients. Sequential samples showed increased bone marrow CD4/CD8 ratios in all cases of progression to bone marrow involvement. Immunohistochemistry showed CD4+, CD57+, programmed death-1+, forkhead box protein 3+ and CD21+ cells accumulated inside the lymphoma infiltrates, whereas CD8+, CD56+ and CD68+ cells were outside the infiltrates. This study provides evidence in vivo that the microenvironment changes upon follicular lymphoma involvement.
Collapse
|
39
|
Fecteau JF, Kipps TJ. Structure and function of the hematopoietic cancer niche: focus on chronic lymphocytic leukemia. Front Biosci (Schol Ed) 2012. [PMID: 22202043 DOI: 10.2741/251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chronic Lymphocytic Leukemia (CLL) is a B cell malignancy characterized by the accumulation of mature monoclonal CD5-positive B cells in the blood, secondary lymphoid tissues, and marrow. The infiltration of CLL cells in lymphoid tissues is a key element of disease pathogenesis. It is in such tissues that are found the microenvironments that provide CLL cells protection from spontaneous and/or drug-induced apoptosis. CLL cells actively shape their microenvironment by producing cytokines and chemokines, and by subverting normal accessory cells to promote leukemia-cell survival, proliferation, and escape from immune detection. In this review, we discuss how CLL cells disrupt the niches required for normal hematopoiesis or immune function and subvert normal cells in the microenvironment to support neoplastic cell growth and survival.
Collapse
Affiliation(s)
- Jessi-F Fecteau
- Rebecca and John Moores Cancer Center, UCSD, La Jolla, CA 92093-0820, USA
| | | |
Collapse
|
40
|
Wang J, Ke XY. The four types of Tregs in malignant lymphomas. J Hematol Oncol 2011; 4:50. [PMID: 22151904 PMCID: PMC3253040 DOI: 10.1186/1756-8722-4-50] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 12/09/2011] [Indexed: 01/29/2023] Open
Abstract
Regulatory T cells (Tregs) are a specialized subpopulation of CD4+ T cells, which act to suppress the activation of other immune cells. Tregs represent important modulators for the interaction between lymphomas and host microenvironment. Lymphomas are a group of serious and frequently fatal malignant diseases of lymphocytes. Recent studies revealed that some lymphoma T cells might adopt a Treg profile. Assessment of Treg phenotypes and genotypes in patients may offer prediction of outcome in many types of lymphomas including diffuse large B-cell lymphoma, follicular lymphoma, cutaneous T cell lymphoma, and Hodgkin's lymphoma. Based on characterized roles of Tregs in lymphomas, we can categorize the various roles into four groups: (a) suppressor Tregs; (b) malignant Tregs; (c) direct tumor-killing Tregs; and (d) incompetent Tregs. The classification into four groups is significant in predicting prognosis and designing Tregs-based immunotherapies for treating lymphomas. In patients with lymphomas where Tregs serve either as suppressor Tregs or malignant Tregs, anti-tumor cytotoxicity is suppressed thus decreased numbers of Tregs are associated with a good prognosis. In contrast, in patients with lymphomas where Tregs serve as tumor-killing Tregs and incompetent Tregs, anti-tumor cytotoxicity is enhanced or anti-autoimmune Tregs activities are weakened thus increased numbers of Tregs are associated with a good prognosis and reduced numbers of Tregs are associated with a poor prognosis. However, the mechanisms underlying the various roles of Tregs in patients with lymphomas remain unknown. Therefore, further research is needed in this regard as well as the utility of Tregs as prognostic factors and therapy strategies in different lymphomas.
Collapse
Affiliation(s)
- Jing Wang
- Department of Hematology and Lymphoma Research Center, Peking University, Third Hospital, (Huayuan North Road 49#), Beijing (100191), PR China
| | | |
Collapse
|
41
|
Hilchey SP, Rosenberg AF, Hyrien O, Secor-Socha S, Cochran MR, Brady MT, Wang JCE, Sanz I, Burack WR, Quataert SA, Bernstein SH. Follicular lymphoma tumor-infiltrating T-helper (T(H)) cells have the same polyfunctional potential as normal nodal T(H) cells despite skewed differentiation. Blood 2011; 118:3591-602. [PMID: 21821704 PMCID: PMC3186335 DOI: 10.1182/blood-2011-03-340646] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 07/23/2011] [Indexed: 12/28/2022] Open
Abstract
The follicular lymphoma (FL) T-cell microenvironment plays a critical role in the biology of this disease. We therefore determined the lineage, differentiation state, and functional potential of FL-infiltrating CD4(+) T-helper cells (T(H)) compared with reactive and normal lymph node (NLN) T(H) cells. Relative to NLNs, FL cells have decreased proportions of naive and central memory but increased proportions of effector memory T(H) cells. We further show differences in the distribution and anatomical localization of CXCR5(+) T(H) populations that, on the basis of transcription factor analysis, include both regulatory and follicular helper T cells. On Staphylococcus enterotoxin-B stimulation, which stimulates T cells through the T-cell receptor, requires no processing by APCs, and can overcome regulator T cell-mediated suppression, the proportion of uncommitted primed precursor cells, as well as T(H)2 and T(H)17 cells is higher in FL cells than in reactive lymph nodes or NLNs. However, the proportion of T(H)1 and polyfunctional T(H) cells (producing multiple cytokines simultaneously) is similar in FL cells and NLNs. These data suggest that, although T(H)-cell differentiation in FL is skewed compared with NLNs, FL T(H) cells should have the same intrinsic ability to elicit antitumor effector responses as NLN T(H) cells when tumor suppressive mechanisms are attenuated.
Collapse
MESH Headings
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cluster Analysis
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Gene Expression Profiling
- Humans
- Immunologic Memory/genetics
- Immunologic Memory/physiology
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/physiology
- Lymphoma, Follicular/genetics
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/metabolism
- Lymphoma, Follicular/pathology
- Microarray Analysis
- Proto-Oncogene Proteins c-bcl-6
- Receptors, CXCR5/genetics
- Receptors, CXCR5/metabolism
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- T-Lymphocytes, Helper-Inducer/physiology
Collapse
Affiliation(s)
- Shannon P Hilchey
- James P. Wilmot Cancer Center, Lymphoma Biology Program, University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Soluble IL-2Rα facilitates IL-2-mediated immune responses and predicts reduced survival in follicular B-cell non-Hodgkin lymphoma. Blood 2011; 118:2809-20. [PMID: 21719603 DOI: 10.1182/blood-2011-03-340885] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Elevated serum levels of the soluble form of IL-2 receptor α (sIL-2Rα) have been correlated with a poor prognosis in a variety of different types of cancers. However, its biologic relevance remains unclear and controversial. In patients with follicular B-cell non-Hodgkin lymphoma (FL), we observed that serum sIL-2Rα levels were elevated compared with controls and that elevated sIL-2Rα levels before treatment were associated with a poor outcome. To explore the mechanism by which sIL-2Rα may contribute to a poor prognosis in FL, we determined the effects of sIL-2Rα on IL-2 signaling and found that the sIL-2Rα-IL-2 complex promoted T-cell differentiation toward to inhibitory T(reg) cells rather than T(H)1 or T(H)17 cells. Shed by activated T cells that express membrane-bound IL-2Rα, sIL-2Rα further enhanced IL-2-mediated phosphorylation of Stat5 thereby significantly up-regulating Foxp3 expression in CD4(+) T cells. We found that CD4(+) T cells treated with either IL-2 or sIL-2Rα-IL-2 complex, but not with sIL-2Rα alone, inhibited the function of CD8(+) T cells. Taken together, these results indicate that sIL-2Rα actually plays an active biologic role in FL by binding IL-2 and promoting IL-2 signaling rather than depleting IL-2 and blocking its function.
Collapse
|
44
|
|
45
|
Role of CD200 expression in regulation of metastasis of EMT6 tumor cells in mice. Breast Cancer Res Treat 2010; 130:49-60. [PMID: 21165772 DOI: 10.1007/s10549-010-1259-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 11/10/2010] [Indexed: 10/18/2022]
Abstract
Previous studies have confirmed that levels of CD200 expression on the cells of the transplantable EMT6 mouse breast cancer line are increased markedly during growth in immunocompetent mice, unlike the persistent low levels of expression observed in NOD-SCID.IL-2(γr-/-) mice or mice with generalized over-expression of a CD200 transgene (CD200(tg) mice). Faster tumor growth occurs in both of these latter mice, with decreased evidence for a host immune reaction in lymph nodes draining the tumor (DLN). We now report evidence for a role for CD200 expression (by the host and/or tumor cells) in increased seeding of tumor cells to DLN in immunocompromised (CD200(tg) or NOD-SCID.IL-2(γr-/-)) vs immunocompetent mice, by limiting dilution cloning of tumor cells from DLN (vs contralateral lymph nodes, CLN), using control and GFP-tagged EMT6 cells. Neutralization of expressed CD200 by anti-CD200mAbs decreased the tumor metastasis at the same time as increasing detection of cytotoxic anti-tumor immune cells in DLN. Infusion of either anti-CD4 to deplete T-effector cells, or anti-TGFβ antibody, increased metastasis to DLN, as did indeed the infusion of EMT6 cells selected for the loss of TGFβRII expression. It is concluded that the increased CD200 expression by breast cancer cells (and/or host tissue) may be an important variable involved in determining the risk of metastasis.
Collapse
|
46
|
Akers NK, Curry JD, Conde L, Bracci PM, Smith MT, Skibola CF. Association of HLA-DQB1 alleles with risk of follicular lymphoma. Leuk Lymphoma 2010; 52:53-8. [PMID: 21133715 DOI: 10.3109/10428194.2010.532888] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In a recent genome-wide association study of follicular lymphoma (FL), we identified novel risk alleles on chromosome 6p21.33 that appeared to be part of an extended haplotype including HLA-DRB1*0101, DQA1*0101, and DQB1*0501. To follow up on these findings, we obtained 2-4 digit HLA-DQB1 allelotypes on a subset of 265 cases of FL and 757 controls using a novel assay that applies multiplexed ligation-dependent probe amplification (MLPA). We confirmed a positive association between FL and the HLA-DQB1*05 allele group (OR = 1.70, 95% CI 1.28-2.27; adjusted p-value = 0.013) and also identified an allele group inversely associated with FL risk, HLA-DQB1*06 (OR = 0.51, 95% CI 0.38-0.69; adjusted p-value = 4.46 × 10(-5)). Although these findings require verification, the role of HLA class II proteins in B-cell survival and proliferation makes this a biologically plausible association.
Collapse
Affiliation(s)
- Nicholas K Akers
- School of Public Health, University of California, Berkeley, CA, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Bruneau J, Canioni D, Renand A, Marafioti T, Paterson JC, Martin-Garcia N, Gaulard P, Delfau MH, Hermine O, Macintyre E, Brousse N, Asnafi V. Regulatory T-cell depletion in angioimmunoblastic T-cell lymphoma. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:570-4. [PMID: 20566750 DOI: 10.2353/ajpath.2010.100150] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is the most frequent nodal T-cell lymphoma and is characterized by a polymorphic lymph node infiltrate, various dysimmune disorders, and a poor prognosis. Regulatory T-cells (Treg) play an emerging role in the prognosis of non-Hodgkin B-cell lymphoma and mediate significant autoreactive T-cell suppression. In this report, we demonstrate that numbers of Treg are significantly decreased in AITL lymph nodes [n = 30, 91 (40-195) per high power fields] compared with follicular lymphoma [n = 19, 179 (86-355)] and reactive lymph nodes [n = 8, 186 (140-265)]. Moreover, the few Treg in lymph nodes of AITL are resting Treg (rTreg) and have a naive CD45RA+, PD1-, and ICOS- phenotype [n = 5, 57% of Treg are CD45RA+ (16-96)], in contrast to the Treg in follicular lymphomas [n = 5, 7.4% (1-13)] or reactive lymph nodes [n = 7, 18.6% (6-48)]. Interestingly, Treg depletion was not observed in AITL peripheral blood at diagnosis. Altogether, these data suggest that Treg depletion could contribute to the nodal neoplastic T(FH) expansion and dysimmune symptoms in AITL.
Collapse
Affiliation(s)
- Julie Bruneau
- Department of Hematology, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris, Paris Descartes University, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Relander T, Johnson NA, Farinha P, Connors JM, Sehn LH, Gascoyne RD. Prognostic Factors in Follicular Lymphoma. J Clin Oncol 2010; 28:2902-13. [DOI: 10.1200/jco.2009.26.1693] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Follicular lymphoma (FL) is one of the most common types of non-Hodgkin's lymphoma. It is usually diagnosed at an advanced stage, for which many treatment options exist, however, no curative standard therapy has been identified. The outcome is highly variable with a median survival of approximately 10 years. The life expectancy of patients with FL has been extended with the use of rituximab, a monoclonal antibody targeting the CD20 antigen on FL cells, but there remains a group of patients who fail to respond to chemoimmunotherapy and die early of their disease. Transformation of FL to an aggressive histology is an important event with high morbidity and mortality. The Follicular Lymphoma International Prognostic Index has become the clinically useful prognostic tool, but gives only a rough estimate of expected outcome. There is a need for useful biomarkers for prediction of the disease course of single patients to individualize therapy, especially in the new era of chemoimmunotherapy.
Collapse
Affiliation(s)
- Thomas Relander
- From the Departments of Pathology & Laboratory Medicine and the Division of Medical Oncology, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Nathalie A. Johnson
- From the Departments of Pathology & Laboratory Medicine and the Division of Medical Oncology, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Pedro Farinha
- From the Departments of Pathology & Laboratory Medicine and the Division of Medical Oncology, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Joseph M. Connors
- From the Departments of Pathology & Laboratory Medicine and the Division of Medical Oncology, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Laurie H. Sehn
- From the Departments of Pathology & Laboratory Medicine and the Division of Medical Oncology, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Randy D. Gascoyne
- From the Departments of Pathology & Laboratory Medicine and the Division of Medical Oncology, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
49
|
Regulatory T Cell as a Target for Cancer Therapy. Arch Immunol Ther Exp (Warsz) 2010; 58:179-90. [DOI: 10.1007/s00005-010-0075-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 12/06/2009] [Indexed: 12/13/2022]
|
50
|
Barnas JL, Simpson-Abelson MR, Yokota SJ, Kelleher RJ, Bankert RB. T cells and stromal fibroblasts in human tumor microenvironments represent potential therapeutic targets. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2010; 3:29-47. [PMID: 21209773 PMCID: PMC2990491 DOI: 10.1007/s12307-010-0044-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 03/02/2010] [Indexed: 12/16/2022]
Abstract
The immune system of cancer patients recognizes tumor-associated antigens expressed on solid tumors and these antigens are able to induce tumor-specific humoral and cellular immune responses. Diverse immunotherapeutic strategies have been used in an attempt to enhance both antibody and T cell responses to tumors. While several tumor vaccination strategies significantly increase the number of tumor-specific lymphocytes in the blood of cancer patients, most vaccinated patients ultimately experience tumor progression. CD4+ and CD8+ T cells with an effector memory phenotype infiltrate human tumor microenvironments, but most are hyporesponsive to stimulation via the T cell receptor (TCR) and CD28 under conditions that activate memory T cells derived from the peripheral blood of the cancer patients or normal donors. Attempts to identify cells and molecules responsible for the TCR signaling arrest of tumor-infiltrating T cells have focused largely upon the immunosuppressive effects of tumor cells, tolerogenic dendritic cells and regulatory T cells. Here we review potential mechanisms by which human T cell function is arrested in the tumor microenvironment with a focus on the immunomodulatory effects of stromal fibroblasts. Determining in vivo which cells and molecules are responsible for the TCR arrest in human tumor-infiltrating T cells will be necessary to formulate and test strategies to prevent or reverse the signaling arrest of the human T cells in situ for a more effective design of tumor vaccines. These questions are now addressable using novel human xenograft models of tumor microenvironments.
Collapse
Affiliation(s)
- Jennifer L. Barnas
- Department of Microbiology and Immunology, Witebsky Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| | - Michelle R. Simpson-Abelson
- Department of Microbiology and Immunology, Witebsky Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| | - Sandra J. Yokota
- Department of Microbiology and Immunology, Witebsky Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| | - Raymond J. Kelleher
- Department of Microbiology and Immunology, Witebsky Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| | - Richard B. Bankert
- Department of Microbiology and Immunology, Witebsky Center, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, 138 Farber Hall, 3435 Main Street, Buffalo, NY 14214 USA
| |
Collapse
|