Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell ; 183(3): 818-834.e13, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33038342

ABSTRACT

Many approaches to identify therapeutically relevant neoantigens couple tumor sequencing with bioinformatic algorithms and inferred rules of tumor epitope immunogenicity. However, there are no reference data to compare these approaches, and the parameters governing tumor epitope immunogenicity remain unclear. Here, we assembled a global consortium wherein each participant predicted immunogenic epitopes from shared tumor sequencing data. 608 epitopes were subsequently assessed for T cell binding in patient-matched samples. By integrating peptide features associated with presentation and recognition, we developed a model of tumor epitope immunogenicity that filtered out 98% of non-immunogenic peptides with a precision above 0.70. Pipelines prioritizing model features had superior performance, and pipeline alterations leveraging them improved prediction performance. These findings were validated in an independent cohort of 310 epitopes prioritized from tumor sequencing data and assessed for T cell binding. This data resource enables identification of parameters underlying effective anti-tumor immunity and is available to the research community.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes/immunology , Neoplasms/immunology , Alleles , Antigen Presentation/immunology , Cohort Studies , Humans , Peptides/immunology , Programmed Cell Death 1 Receptor , Reproducibility of Results
3.
Nat Rev Immunol ; 3(6): 463-71, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12776206

ABSTRACT

The completion of the genome sequences of both humans and mice challenges biologists to determine gene function on a vast, whole-organism scale. Both phenotype-based ('forward') and gene-based ('reverse') strategies are being developed to approach this issue. Forward-genetic approaches, however, provide the unique ability of assigning function to genes in an unbiased, global manner that is independent of previous assumptions about gene function. In this article, we compare various genetic technologies for their potential role in dissecting immune-system development and function, with particular emphasis on the worldwide efforts that use chemical mutagenesis as a forward-genetic strategy.


Subject(s)
Genes/physiology , Genome, Human , Immune System/physiology , Phenotype , Animals , Ethylnitrosourea , Humans , Mice , Mice, Mutant Strains , Mutagenesis, Site-Directed
5.
Front Immunol ; 14: 1166135, 2023.
Article in English | MEDLINE | ID: mdl-37153574

ABSTRACT

Regulatory T (Treg) cells are essential for maintaining peripheral tolerance, preventing autoimmunity, and limiting chronic inflammatory diseases. This small CD4+ T cell population can develop in the thymus and in the peripheral tissues of the immune system through the expression of an epigenetically stabilized transcription factor, FOXP3. Treg cells mediate their tolerogenic effects using multiple modes of action, including the production of inhibitory cytokines, cytokine starvation of T effector (e.g., IL-2), Teff suppression by metabolic disruption, and modulation of antigen-presenting cell maturation or function. These activities together result in the broad control of various immune cell subsets, leading to the suppression of cell activation/expansion and effector functions. Moreover, these cells can facilitate tissue repair to complement their suppressive effects. In recent years, there has been an effort to harness Treg cells as a new therapeutic approach to treat autoimmune and other immunological diseases and, importantly, to re-establish tolerance. Recent synthetic biological advances have enabled the cells to be genetically engineered to achieve tolerance and antigen-specific immune suppression by increasing their specific activity, stability, and efficacy. These cells are now being tested in clinical trials. In this review, we highlight both the advances and the challenges in this arena, focusing on the efforts to develop this new pillar of medicine to treat and cure a variety of diseases.


Subject(s)
Immune System Diseases , T-Lymphocytes, Regulatory , Humans , CD4-Positive T-Lymphocytes , Immune Tolerance , Immunosuppression Therapy , Immune System Diseases/metabolism , Cytokines/metabolism
6.
Eur J Immunol ; 41(4): 902-15, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21416464

ABSTRACT

Members of the CD28 family play important roles in regulating T-cell functions and share a common gene structure profile. We have identified VSTM3 as a protein whose gene structure matches that of the other CD28 family members. This protein (also known as TIGIT and WUCAM) has been previously shown to affect immune responses and is expressed on NK cells, activated and memory T cells, and Tregs. The nectin-family proteins CD155 and CD112 serve as counter-structures for VSTM3, and CD155 and CD112 also bind to the activating receptor CD226 on T cells and NK cells. Hence, this group of interacting proteins forms a network of molecules similar to the well-characterized CD28-CTLA-4-CD80-CD86 network. In the same way that soluble CTLA-4 can be used to block T-cell responses, we show that soluble Vstm3 attenuates T-cell responses in vitro and in vivo. Moreover, animals deficient in Vstm3 are more sensitive to autoimmune challenges indicating that this new member of the CD28 family is an important regulator of T-cell responses.


Subject(s)
CD28 Antigens/immunology , Receptors, Immunologic/immunology , T-Lymphocytes/immunology , Animals , Autoimmune Diseases/immunology , Cells, Cultured , Dendritic Cells/immunology , Humans , Mice , Rats , Receptors, Immunologic/deficiency , T-Lymphocytes/chemistry
7.
Genome Med ; 10(1): 79, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30376867

ABSTRACT

For at least 300 years the immune system has been targeted to improve human health. Decades of work advancing immunotherapies against infection and autoimmunity paved the way for the current explosion in cancer immunotherapies. Pathways targeted for therapeutic intervention in autoimmune diseases can be modulated in the opposite sense in malignancy and infectious disease. We discuss the basic principles of the immune response, how these are co-opted in chronic infection and malignancy, and how these can be harnessed to treat disease. T cells are at the center of immunotherapy. We consider the complexity of T cell functional subsets, differentiation states, and extrinsic and intrinsic influences in the design, success, and lessons from immunotherapies. The integral role of checkpoints in the immune response is highlighted by the rapid advances in FDA approvals and the use of therapeutics that target the CTLA-4 and PD-1/PD-L1 pathways. We discuss the distinct and overlapping mechanisms of CTLA-4 and PD-1 and how these can be translated to combination immunotherapy treatments. Finally, we discuss how the successes and challenges in cancer immunotherapies, such as the collateral damage of immune-related adverse events following checkpoint inhibition, are informing treatment of autoimmunity, infection, and malignancy.


Subject(s)
Autoimmunity , Immunotherapy , Infections/immunology , Infections/therapy , Neoplasms/immunology , Neoplasms/therapy , Humans , T-Lymphocytes/immunology
8.
Curr Opin Immunol ; 15(6): 718-24, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14630208

ABSTRACT

The identification of two transcription factors that, when mutated, are responsible for severe autoimmune disease in humans is leading to a better understanding of the fundamental processes involved in T-cell tolerance. Both AIRE and FOXP3, identified initially via their association with genetically manipulated mice, are critically involved in tolerance induction in humans. Although mutations in these genes may cause rare but serious diseases, it is likely that other transcription factors will contribute to the genetic load that predisposes certain individuals to disease.


Subject(s)
Autoimmunity/physiology , Transcription Factors/physiology , Animals , Autoimmunity/genetics , CD4-Positive T-Lymphocytes/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Forkhead Transcription Factors , Mice , Receptors, Interleukin-2/physiology , Thymus Gland/physiology , Transcription Factors/genetics , AIRE Protein
9.
Nat Rev Immunol ; 14(5): 343-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24722479

ABSTRACT

It has been 65 years since the scurfy mutation arose spontaneously in mice at the Oak Ridge National Laboratory in the United States, and it is 13 years since the molecular cloning of the forkhead box P3 (FOXP3) gene was reported. In this Timeline article, we review the events that have occurred during and since this time. This is not meant as an exhaustive review of the biology of FOXP3 or of regulatory T cells, rather it is an attempt to highlight the landmark events that have demonstrated the importance of FOXP3 in immune function. These events have driven, and continue to drive, the extensive research effort to fully understand the role of regulatory T cells in the immune system.


Subject(s)
Forkhead Transcription Factors/immunology , Animals , Humans , Immune System/immunology , T-Lymphocytes, Regulatory/immunology
10.
J Exp Med ; 205(12): 2899-913, 2008 Nov 24.
Article in English | MEDLINE | ID: mdl-19015308

ABSTRACT

Hem1 (Hematopoietic protein 1) is a hematopoietic cell-specific member of the Hem family of cytoplasmic adaptor proteins. Orthologues of Hem1 in Dictyostelium discoideum, Drosophila melanogaster, and Caenorhabditis elegans are essential for cytoskeletal reorganization, embryonic cell migration, and morphogenesis. However, the in vivo functions of mammalian Hem1 are not known. Using a chemical mutagenesis strategy in mice to identify novel genes involved in immune cell functions, we positionally cloned a nonsense mutation in the Hem1 gene. Hem1 deficiency results in defective F-actin polymerization and actin capping in lymphocytes and neutrophils caused by loss of the Rac-controlled actin-regulatory WAVE protein complex. T cell development is disrupted in Hem1-deficient mice at the CD4(-)CD8(-) (double negative) to CD4(+)CD8(+) (double positive) cell stages, whereas T cell activation and adhesion are impaired. Hem1-deficient neutrophils fail to migrate in response to chemotactic agents and are deficient in their ability to phagocytose bacteria. Remarkably, some Rac-dependent functions, such as Th1 differentiation and nuclear factor kappaB (NF-kappaB)-dependent transcription of proinflammatory cytokines proceed normally in Hem1-deficient mice, whereas the production of Th17 cells are enhanced. These results demonstrate that Hem1 is essential for hematopoietic cell development, function, and homeostasis by controlling a distinct pathway leading to cytoskeletal reorganization, whereas NF-kappaB-dependent transcription proceeds independently of Hem1 and F-actin polymerization.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Immunity, Innate/physiology , Lymphopoiesis/physiology , Membrane Proteins , Point Mutation , Actins/metabolism , Anemia/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Movement/physiology , DNA Mutational Analysis , Hematopoietic Stem Cells/physiology , Hematopoietic System/cytology , Hematopoietic System/physiology , Interferon-gamma/immunology , Interleukin-17/metabolism , Interleukin-2/immunology , Lymphocyte Activation , Lymphopenia/immunology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Neutrophils/immunology , Neutrophils/physiology , Phagocytosis/physiology , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Transplantation Chimera
11.
Curr Protoc Immunol ; Chapter 3: Unit 3.18, 2006 Mar.
Article in English | MEDLINE | ID: mdl-18432973

ABSTRACT

Most T cell development occurs within the thymus and includes a series of selection processes that are, in large part, still poorly understood. Studies of T cell development have been greatly advanced by the description of multiple phenotypic subsets of T cells and their maturational relationships. This unit describes a system for observing and modulating T cell development in vitro via the culture of entire mouse fetal thymic lobes. Methods are included for the isolation of fetal thymi and culture to allow for normal T cell development on either transwell plates or Gelfoam sponges. A method for depleting hematopoietic cells from thymic lobes using 2'-deoxyguanosine and subsequent reconstitution with precursor cells is also described. This protocol is valuable for the study of tolerance and T cell selection. A support protocol describing methods of altering and monitoring T cell development are outlined. In addition, methods for culturing fetal thymic lobes under high oxygen submersion conditions and for the preparation of reaggregate thymus organ cultures are provided. Finally, a simple and practical method that allows for the thymus-independent generation of T cells from defined sources of stem/progenitor cells by OP9-DL1 coculture is described.


Subject(s)
Coculture Techniques/methods , Lymphopoiesis , Organ Culture Techniques/methods , T-Lymphocytes/physiology , Thymus Gland/embryology , Adipocytes/cytology , Animals , Cell Differentiation , Fetus , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/physiology , Mice , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/physiology
12.
Immunology ; 119(2): 203-11, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17005002

ABSTRACT

FOXP3 has been identified as a key regulator of immune homeostasis. Mutations within the FOXP3 gene result in dysregulated CD4+ T-cell function and elevated cytokine production, leading to lymphoproliferative disease. FOXP3 expression in CD4+ T cells is primarily detected with the CD4+ CD25+ regulatory T-cell population. In humans the protein is detected as a doublet following immunoblot analysis. The lower band of the doublet has been identified as a splice isoform lacking a region corresponding to exon 2. The aim of this study was to investigate whether the splice variant form lacking exon 2 and a new novel splice variant lacking both exons 2 and 7, were functional inhibitors of CD4+ T-cell activation. The data generated showed that full-length FOXP3 and both splice variant forms of the protein were functional repressors of CD4+ T-cell activation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/genetics , Lymphocyte Activation/genetics , RNA Splice Sites/genetics , Base Sequence , CD28 Antigens/immunology , Cell Proliferation , Cells, Cultured , Cloning, Molecular , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Molecular Sequence Data , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Antigen, T-Cell/immunology , Sequence Alignment , Transfection
13.
J Immunol ; 177(9): 5852-60, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17056509

ABSTRACT

CD4+CD25+ regulatory T cells can prevent and resolve intestinal inflammation in the murine T cell transfer model of colitis. Using Foxp3 as a marker of regulatory T cell activity, we now provide a comprehensive analysis of the in vivo distribution of Foxp3+CD4+CD25+ cells in wild-type mice, and during cure of experimental colitis. In both cases, Foxp3+CD4+CD25+ cells were found to accumulate in the colon and secondary lymphoid organs. Importantly, Foxp3+ cells were present at increased density in colon samples from patients with ulcerative colitis or Crohn's disease, suggesting similarities in the behavior of murine and human regulatory cells under inflammatory conditions. Cure of murine colitis was dependent on the presence of IL-10, and IL-10-producing CD4+CD25+ T cells were enriched within the colon during cure of colitis and also under steady state conditions. Our data indicate that although CD4+CD25+ T cells expressing Foxp3 are present within both lymphoid organs and the colon, subsets of IL-10-producing CD4+CD25+ T cells are present mainly within the intestinal lamina propria suggesting compartmentalization of the regulatory T cell response at effector sites.


Subject(s)
Colitis/immunology , Colitis/therapy , Forkhead Transcription Factors/metabolism , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Animals , CD4 Antigens/analysis , Colitis/pathology , Colon/chemistry , Colon/immunology , Colon/pathology , Disease Models, Animal , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/genetics , Humans , Interleukin-10/analysis , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/analysis , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Knockout , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/chemistry
14.
Eur J Immunol ; 35(12): 3424-32, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16285010

ABSTRACT

The forkhead-family transcription factor FoxP3 is important for the development and function of CD4+CD25+ regulatory T cells. While the overall phenotypic effects of FoxP3 expression are evident, the mechanism by which FoxP3 regulates T cell activation is not well understood. CD4+ T cells from mice that express a FoxP3 Tg are refractory to TCR-mediated stimulation, failing to proliferate or produce cytokines, but possess suppressive activity towards normal T cells. In this report we show that these T cells express elevated levels of mRNA for pro-apoptotic genes and undergo rapid apoptosis following stimulation. These T cells also display slower cell cycle transit following activation, suggesting that FoxP3 is capable of regulating the ability of T cells to respond to TCR-mediated activation. Lastly, we show that contrary to expected results, under Th1 or Th2 driving conditions, CD4+ T cells from FoxP3 Tg mice differentiate into effector cells. Concomitant with differentiation is a loss of FoxP3 mRNA and protein. These data demonstrate that FoxP3 levels regulate T cell function, and that FoxP3 itself is dynamically regulated during effector T cell differentiation.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation/immunology , Lymphocyte Activation/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Forkhead Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/biosynthesis , Receptors, Antigen, T-Cell/physiology
15.
Immunity ; 19(2): 165-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12932350

ABSTRACT

The recent characterization of a mutant strain of mice generated five decades ago in a program to study ionizing radiation in mammals (and, ironically, derived from a nonmutagenized control animal) is helping to dissect a now resurgent area of immunology. Despite a vast literature during the 1970s, the study of suppressor T cells had been largely abandoned until the publication of several seminal papers rekindled interest in what are today generally referred to as regulatory T (TR) cells. The identification of the transcription factor Foxp3 as the gene responsible for the defect in scurfy mice, and subsequently, the demonstration of its critical involvement in the generation of TR cells, provides an important molecular insight into this essential cell lineage.


Subject(s)
DNA-Binding Proteins/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors , Immune Tolerance , Mice , Models, Immunological , Mutation , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
16.
Nat Immunol ; 4(4): 337-42, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12612581

ABSTRACT

The molecular properties that characterize CD4+CD25+ regulatory T cells (TR cells) remain elusive. Absence of the transcription factor Scurfin (also known as forkhead box P3 and encoded by Foxp3) causes a rapidly fatal lymphoproliferative disease, similar to that seen in mice lacking cytolytic T lymphocyte-associated antigen 4 (CTLA-4). Here we show that Foxp3 is highly expressed by T(R) cells and is associated with T(R) cell activity and phenotype. Scurfin-deficient mice lack T(R) cells, whereas mice that overexpress Foxp3 possess more T(R) cells. In Foxp3-overexpressing mice, both CD4+CD25- and CD4-CD8+ T cells show suppressive activity and CD4+CD25- cells express glucocorticoid-induced tumor-necrosis factor receptor-related (GITR) protein. The forced expression of Foxp3 also delays disease in CTLA-4-/- mice, indicating that the Scurfin and CTLA-4 pathways may intersect and providing further insight into the T(R) cell lineage.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Immunoconjugates , Receptors, Interleukin-2/metabolism , Abatacept , Animals , Antigens, CD , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , DNA-Binding Proteins/genetics , Forkhead Transcription Factors , Glucocorticoid-Induced TNFR-Related Protein , Interleukin-10/metabolism , Mice , Mice, Transgenic , Receptors, Interleukin-2/immunology , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Transforming Growth Factor beta/metabolism
17.
Int Immunol ; 14(9): 1039-53, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12202401

ABSTRACT

As a result of a transgene insertion and chromosomal deletion, a mutant mouse strain has been found that is defective in the lineage commitment step that produces a balance of alphabeta and gammadelta T cells. The mice produce a reduced population of alphabeta CD4 T cells and almost no alphabeta CD8 T cells. Within the CD4 and CD8 populations in the thymus there exist abnormal subsets that express the gammadelta TCR. These gammadelta TCR-expressing cells populate the peripheral lymphoid organs such that up to 75% of the CD8 T cells in the lymph nodes and spleen express a gammadelta TCR. Further analyses indicate that the regulation that prevents dual TCR expression has been impaired. The locus of insertion and deletion was mapped to chromosome 10 26 cM. We have analyzed the entire locus and, in addition, the gene expression changes in early thymocytes were analyzed by gene array technology. The analysis of this mutant strain indicates that the alphabeta versus gammadelta lineage decision can be profoundly disregulated independently of successful gene rearrangements.


Subject(s)
Adaptor Proteins, Signal Transducing , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Carrier Proteins/genetics , Integrases/genetics , Mice , Mice, Transgenic , Promoter Regions, Genetic , Thymus Gland/abnormalities , Thymus Gland/growth & development , Viral Proteins/genetics
18.
Bioorg Med Chem Lett ; 12(7): 1093-7, 2002 Apr 08.
Article in English | MEDLINE | ID: mdl-11909725

ABSTRACT

A novel series of TNF-alpha inhibitors based on a benzobicyclooctane scaffold is reported. The compounds display good potency in inhibiting TNF-alpha induced apoptosis and NF kappa B activation. Additionally, they are selective for TNF-alpha as they do not inhibit apoptosis induced by soluble Fas ligand. The compounds described here can act as leads for future medicinal chemistry efforts and may also be useful tools for elucidating the TNF-alpha signaling pathway.


Subject(s)
Apoptosis/drug effects , Octanes/pharmacology , Signal Transduction/physiology , Tumor Cells, Cultured/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Apoptosis/physiology , Fas Ligand Protein , Humans , Lung Neoplasms/drug therapy , Membrane Glycoproteins/pharmacology , Molecular Structure , NF-kappa B/drug effects , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
19.
J Immunol ; 173(5): 2995-3001, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15322158

ABSTRACT

Using a mouse mutagenesis screen, we have identified CD83 as being critical for the development of CD4(+) T cells and for their function postactivation. CD11c(+) dendritic cells develop and function normally in mice with a mutated CD83 gene but CD4(+) T cell development is substantially reduced. Additionally, we now show that those CD4(+) cells that develop in a CD83 mutant animal fail to respond normally following allogeneic stimulation. This is at least in part due to an altered cytokine expression pattern characterized by an increased production of IL-4 and IL-10 and diminished IL-2 production. Thus, in addition to its role in selection of CD4(+) T cells, absence of CD83 results in the generation of cells with an altered activation and cytokine profile.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunoglobulins/genetics , Membrane Glycoproteins/genetics , Amino Acid Sequence , Animals , Antigens, CD , Base Sequence , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Dendritic Cells/immunology , Female , Immunoglobulins/immunology , Male , Membrane Glycoproteins/immunology , Mice , Molecular Sequence Data , Mutation , Pedigree , CD83 Antigen
SELECTION OF CITATIONS
SEARCH DETAIL