Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 1016179, 2022.
Article in English | MEDLINE | ID: mdl-36569945

ABSTRACT

The optimal use of many biotherapeutics is restricted by Anti-drug antibodies (ADAs) and hypersensitivity responses which can affect potency and ability to administer a treatment. Here we demonstrate that Re-surfacing can be utilized as a generalizable approach to engineer proteins with extensive surface residue modifications in order to avoid binding by pre-existing ADAs. This technique was applied to E. coli Asparaginase (ASN) to produce functional mutants with up to 58 substitutions resulting in direct modification of 35% of surface residues. Re-surfaced ASNs exhibited significantly reduced binding to murine, rabbit and human polyclonal ADAs, with a negative correlation observed between binding and mutational distance from the native protein. Reductions in ADA binding correlated with diminished hypersensitivity responses in an in vivo mouse model. By using computational design approaches to traverse extended distances in mutational space while maintaining function, protein Re-surfacing may provide a means to generate novel or second line therapies for life-saving drugs with limited therapeutic alternatives.


Subject(s)
Asparaginase , Escherichia coli , Humans , Animals , Mice , Rabbits , Asparaginase/genetics , Asparaginase/therapeutic use , Escherichia coli/genetics , Antibodies , Membrane Proteins
2.
Nature ; 598(7882): 662-666, 2021 10.
Article in English | MEDLINE | ID: mdl-34616044

ABSTRACT

The availability of L-arginine in tumours is a key determinant of an efficient anti-tumour T cell response1-4. Consequently, increases of typically low L-arginine concentrations within the tumour may greatly potentiate the anti-tumour responses of immune checkpoint inhibitors, such as programmed death-ligand 1 (PD-L1)-blocking antibodies5. However, currently no means are available to locally increase intratumoural L-arginine levels. Here we used a synthetic biology approach to develop an engineered probiotic Escherichia coli Nissle 1917 strain that colonizes tumours and continuously converts ammonia, a metabolic waste product that accumulates in tumours6, to L-arginine. Colonization of tumours with these bacteria increased intratumoural L-arginine concentrations, increased the number of tumour-infiltrating T cells and had marked synergistic effects with PD-L1 blocking antibodies in the clearance of tumours. The anti-tumour effect of these bacteria was mediated by L-arginine and was dependent on T cells. These results show that engineered microbial therapies enable metabolic modulation of the tumour microenvironment leading to enhanced efficacy of immunotherapies.


Subject(s)
Immunotherapy/methods , Metabolic Engineering , Microorganisms, Genetically-Modified , Neoplasms, Experimental/therapy , Adoptive Transfer , Animals , Arginine/metabolism , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Escherichia coli , Female , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/microbiology , Probiotics , Proteome , Synthetic Biology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
3.
Nat Commun ; 11(1): 2739, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32483165

ABSTRACT

Synthetic biology is a powerful tool to create therapeutics which can be rationally designed to enable unique and combinatorial functionalities. Here we utilize non-pathogenic E coli Nissle as a versatile platform for the development of a living biotherapeutic for the treatment of cancer. The engineered bacterial strain, referred to as SYNB1891, targets STING-activation to phagocytic antigen-presenting cells (APCs) in the tumor and activates complementary innate immune pathways. SYNB1891 treatment results in efficacious antitumor immunity with the formation of immunological memory in murine tumor models and robust activation of human APCs. SYNB1891 is designed to meet manufacturability and regulatory requirements with built in biocontainment features which do not compromise its efficacy. This work provides a roadmap for the development of future therapeutics and demonstrates the transformative potential of synthetic biology for the treatment of human disease when drug development criteria are incorporated into the design process for a living medicine.


Subject(s)
Escherichia coli/immunology , Immunotherapy/methods , Membrane Proteins/immunology , Neoplasms/therapy , Signal Transduction/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Engineering/methods , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Phagocytes/immunology , Phagocytes/metabolism , Signal Transduction/genetics , Synthetic Biology/methods , Synthetic Biology/trends
4.
J Immunol ; 202(9): 2628-2635, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30902900

ABSTRACT

Deletion of CD8+ T cells by dendritic cells (DCs) is recognized as a critical mechanism of immune tolerance to self-antigens. Although DC-mediated peripheral deletion of autoreactive CD8+ T cells has been demonstrated using T cells reactive to model Ags, its role in shaping the naturally occurring polyclonal CD8+ T cell repertoire has not been defined. Using Batf3-/- mice lacking cross-presenting CD8α+ and CD103+ DCs (also known as type 1 conventional [cDC1]), we demonstrate that peripheral deletion of CD8+ T cells reactive to a model tissue Ag is dependent on cDC1. However, endogenous CD8+ T cells from the periphery of Batf3-/- mice do not exhibit heightened self-reactivity, and deep TCR sequencing of CD8+ T cells from Batf3-/- and Batf3+/+ mice reveals that cDC1 have a minimal impact on shaping the peripheral CD8+ T cell repertoire. Thus, although evident in reductionist systems, deletion of polyclonal self-specific CD8+ T cells by cDC1 plays a negligible role in enforcing tolerance to natural self-ligands.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immune Tolerance , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Mice , Mice, Knockout , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Repressor Proteins/genetics , Repressor Proteins/immunology
5.
Immunity ; 44(5): 1102-13, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27130899

ABSTRACT

The promiscuous expression of tissue-restricted antigens in the thymus, driven in part by autoimmune regulator (Aire), is critical for the protection of peripheral tissues from autoimmune attack. Aire-dependent processes are thought to promote both clonal deletion and the development of Foxp3(+) regulatory T (Treg) cells, suggesting that autoimmunity associated with Aire deficiency results from two failed tolerance mechanisms. Here, examination of autoimmune lesions in Aire(-/-) mice revealed an unexpected third possibility. We found that the predominant conventional T cell clonotypes infiltrating target lesions express antigen receptors that were preferentially expressed by Foxp3(+) Treg cells in Aire(+/+) mice. Thus, Aire enforces immune tolerance by ensuring that distinct autoreactive T cell specificities differentiate into the Treg cell lineage; dysregulation of this process results in the diversion of Treg cell-biased clonotypes into pathogenic conventional T cells.


Subject(s)
Autoimmunity , Immune Tolerance , Prostate/immunology , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Regulatory/physiology , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , Autoantigens/immunology , Autoimmunity/genetics , Cell Differentiation , Cell Lineage , Clonal Deletion , Clonal Selection, Antigen-Mediated , Clone Cells , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Immune Tolerance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Cell Antigen Receptor Specificity , Transcription Factors/genetics , AIRE Protein
6.
Immunity ; 44(4): 847-59, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27037189

ABSTRACT

Although antigen recognition mediated by the T cell receptor (TCR) influences many facets of Foxp3(+) regulatory T (Treg) cell biology, including development and function, the cell types that present antigen to Treg cells in vivo remain largely undefined. By tracking a clonal population of Aire-dependent, prostate-specific Treg cells in mice, we demonstrated an essential role for dendritic cells (DCs) in regulating organ-specific Treg cell biology. We have shown that the thymic development of prostate-specific Treg cells required antigen presentation by DCs. Moreover, Batf3-dependent CD8α(+) DCs were dispensable for the development of this clonotype and had negligible impact on the polyclonal Treg cell repertoire. In the periphery, CCR7-dependent migratory DCs coordinated the activation of organ-specific Treg cells in the prostate-draining lymph nodes. Our results demonstrate that the development and peripheral regulation of organ-specific Treg cells are dependent on antigen presentation by DCs, implicating DCs as key mediators of organ-specific immune tolerance.


Subject(s)
Antigen Presentation/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Dendritic Cells/immunology , Prostate/immunology , Repressor Proteins/genetics , Self Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B7-1 Antigen/biosynthesis , B7-1 Antigen/genetics , B7-2 Antigen/biosynthesis , B7-2 Antigen/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CD8 Antigens/metabolism , Cell Differentiation/immunology , Cell Movement/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prostate/cytology , Receptors, Antigen, T-Cell/immunology , Receptors, CCR7/metabolism , Repressor Proteins/immunology , T-Lymphocytes, Regulatory/cytology , Transcription Factors/metabolism , AIRE Protein
7.
Immunol Rev ; 259(1): 245-58, 2014 May.
Article in English | MEDLINE | ID: mdl-24712470

ABSTRACT

Many tumors express antigens that can be specifically or selectively recognized by T lymphocytes, suggesting that T-cell-mediated immunity may be harnessed for the immunotherapy of cancer. However, since tumors originate from normal cells and evolve within the context of self-tissues, the immune mechanisms that prevent the autoimmune attack of normal tissues function in parallel to restrict anti-tumor immunity. In particular, the purging of autoreactive T cells and the development of immune-suppressive regulatory T cells (Tregs) are thought to be major barriers impeding anti-tumor immune responses. Here, we discuss current understanding regarding the antigens recognized by tumor-infiltrating T-cell populations, the mechanisms that shape the repertoire of these cells, and the role of the transcription factor autoimmune regulator (Aire) in these processes. Further elucidation of these principles is likely to be critical for optimizing emerging cancer immunotherapies, and for the rational design of novel therapies exhibiting robust anti-tumor activity with limited toxicity.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , Autoimmunity/genetics , Autoimmunity/immunology , Clonal Selection, Antigen-Mediated/genetics , Clonal Selection, Antigen-Mediated/immunology , Epitopes, T-Lymphocyte , Female , Humans , Immune Tolerance , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Mice , Neoplasms/genetics , Neoplasms/metabolism , Prostate/immunology , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Microenvironment/immunology , AIRE Protein
8.
Oncoimmunology ; 2(7): e24898, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-24073374

ABSTRACT

Little is known about the relative contributions of self-specific regulatory T cells (Tregs) of thymic origin and induced Tregs generated extrathymically to the pool of tumor-infiltrating Tregs. We have recently demonstrated that thymic-derived Tregs reactive to a prostate-associated self antigen are highly and recurrently enriched within oncogene-driven murine prostate cancers.

9.
Science ; 339(6124): 1219-24, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23471412

ABSTRACT

Despite considerable interest in the modulation of tumor-associated Foxp3(+) regulatory T cells (T(regs)) for therapeutic benefit, little is known about the developmental origins of these cells and the nature of the antigens that they recognize. We identified an endogenous population of antigen-specific T(regs) (termed MJ23 T(regs)) found recurrently enriched in the tumors of mice with oncogene-driven prostate cancer. MJ23 T(regs) were not reactive to a tumor-specific antigen but instead recognized a prostate-associated antigen that was present in tumor-free mice. MJ23 T(regs) underwent autoimmune regulator (Aire)-dependent thymic development in both male and female mice. Thus, Aire-mediated expression of peripheral tissue antigens drives the thymic development of a subset of organ-specific T(regs), which are likely coopted by tumors developing within the associated organ.


Subject(s)
Immune Tolerance , Prostate/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/growth & development , Thymus Gland/immunology , Transcription Factors/immunology , Animals , Antigens, Polyomavirus Transforming/genetics , Autoantigens/immunology , CD4 Antigens/analysis , Female , Forkhead Transcription Factors/analysis , Homeodomain Proteins/genetics , Male , Mice , Mice, Transgenic , Prostate-Specific Antigen/immunology , Transcription Factors/genetics , AIRE Protein
10.
Trends Immunol ; 34(1): 33-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22999714

ABSTRACT

Due to the critical role of forkhead box (Fox)p3(+) regulatory T cells (Tregs) in the regulation of immunity and the enrichment of Tregs within many human tumors, several emerging therapeutic strategies for cancer involve the depletion or modulation of Tregs, with the aim of eliciting enhanced antitumor immune responses. Here, we review recent advances in understanding of the fundamental biology of Tregs, and discuss the implications of these findings for current models of tumor-associated Treg biology. In particular, we discuss the context-dependent functional diversity of Tregs, the developmental origins of these cells, and the nature of the antigens that they recognize within the tumor environment. In addition, we highlight critical areas of focus for future research.


Subject(s)
Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/immunology , Cell Lineage , Cell Movement , Humans , Immunotherapy , Neoplasms/therapy , T-Lymphocytes, Regulatory/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...