Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Blood ; 143(6): 507-521, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38048594

ABSTRACT

ABSTRACT: Chimeric antigen receptor T-cell (CAR T) therapy has produced remarkable clinical responses in B-cell neoplasms. However, many challenges limit this class of agents for the treatment of other cancer types, in particular the lack of tumor-selective antigens for solid tumors and other hematological malignancies, such as acute myeloid leukemia (AML), which may be addressed without significant risk of severe toxicities while providing sufficient abundance for efficient tumor suppression. One approach to overcome this hurdle is dual targeting by an antibody-T-cell receptor (AbTCR) and a chimeric costimulatory signaling receptor (CSR) to 2 different antigens, in which both antigens are found together on the cancer cells but not together on normal cells. To explore this proof of concept in AML, we engineered a new T-cell format targeting Wilms tumor 1 protein (WT1) and CD33; both are highly expressed on most AML cells. Using an AbTCR comprising a newly developed TCR-mimic monoclonal antibody against the WT1 RMFPNAPYL (RMF) epitope/HLA-A2 complex, ESK2, and a secondary CSR comprising a single-chain variable fragment directed to CD33 linked to a truncated CD28 costimulatory fragment, this unique platform confers specific T-cell cytotoxicity to the AML cells while sparing healthy hematopoietic cells, including CD33+ myelomonocytic normal cells. These data suggest that this new platform, named AbTCR-CSR, through the combination of a AbTCR CAR and CSR could be an effective strategy to reduce toxicity and improve specificity and clinical outcomes in adoptive T-cell therapy in AML.


Subject(s)
Leukemia, Myeloid, Acute , Single-Chain Antibodies , Humans , T-Lymphocytes , Receptors, Antigen, T-Cell , Leukemia, Myeloid, Acute/pathology , Immunotherapy, Adoptive
2.
Nat Chem Biol ; 18(2): 216-225, 2022 02.
Article in English | MEDLINE | ID: mdl-34969970

ABSTRACT

Chimeric antigen receptor (CAR)-T cells represent a major breakthrough in cancer therapy, wherein a patient's own T cells are engineered to recognize a tumor antigen, resulting in activation of a local cytotoxic immune response. However, CAR-T cell therapies are currently limited to the treatment of B cell cancers and their effectiveness is hindered by resistance from antigen-negative tumor cells, immunosuppression in the tumor microenvironment, eventual exhaustion of T cell immunologic functions and frequent severe toxicities. To overcome these problems, we have developed a novel class of CAR-T cells engineered to express an enzyme that activates a systemically administered small-molecule prodrug in situ at a tumor site. We show that these synthetic enzyme-armed killer (SEAKER) cells exhibit enhanced anticancer activity with small-molecule prodrugs, both in vitro and in vivo in mouse tumor models. This modular platform enables combined targeting of cellular and small-molecule therapies to treat cancers and potentially a variety of other diseases.


Subject(s)
Antineoplastic Agents/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Drug Delivery Systems , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Neoplasms, Experimental , Prodrugs , Receptors, Chimeric Antigen , T-Lymphocytes , Tumor Microenvironment , Xenograft Model Antitumor Assays
3.
J Biol Chem ; 297(5): 101277, 2021 11.
Article in English | MEDLINE | ID: mdl-34619148

ABSTRACT

Nucleic acid-sensing pathways play critical roles in innate immune activation through the production of type I interferon (IFN-I) and proinflammatory cytokines. These factors are required for effective antitumor immune responses. Pharmacological modulators of the pre-mRNA spliceosome splicing factor 3b subunit 1 (SF3B1) are under clinical investigation as cancer cytotoxic agents. However, potential roles of these agents in aberrant RNA generation and subsequent RNA-sensing pathway activation have not been studied. In this study, we observed that SF3B1 pharmacological modulation using pladienolide B (Plad B) induces production of aberrant RNA species and robust IFN-I responses via engagement of the dsRNA sensor retinoic acid-inducible gene I (RIG-I) and downstream interferon regulatory factor 3. We found that Plad B synergized with canonical RIG-I agonism to induce the IFN-I response. In addition, Plad B induced NF-κB responses and secretion of proinflammatory cytokines and chemokines. Finally, we showed that cancer cells bearing the hotspot SF3B1K700E mutation, which leads to global aberrant splicing, had enhanced IFN-I response to canonical RIG-I agonism. Together, these results demonstrate that pharmacological modulation of SF3B1 in cancer cells can induce an enhanced IFN-I response dependent on RIG-I expression. The study suggests that spliceosome modulation may not only induce direct cancer cell cytotoxicity but also initiate an innate immune response via activation of RNA-sensing pathways.


Subject(s)
DEAD Box Protein 58/metabolism , Interferon Type I/metabolism , Phosphoproteins/metabolism , RNA Precursors/metabolism , RNA Splicing Factors/metabolism , Receptors, Immunologic/metabolism , Spliceosomes/metabolism , A549 Cells , Amino Acid Substitution , Animals , DEAD Box Protein 58/genetics , Humans , Interferon Type I/genetics , Mice , Mutation, Missense , Phosphoproteins/genetics , RNA Precursors/genetics , RNA Splicing Factors/genetics , Receptors, Immunologic/genetics , Spliceosomes/genetics , THP-1 Cells
4.
Cancer Immunol Res ; 8(5): 672-684, 2020 05.
Article in English | MEDLINE | ID: mdl-32184297

ABSTRACT

T-cell receptor (TCR)-based therapeutic cells and agents have emerged as a new class of effective cancer therapies. These therapies work on cells that express intracellular cancer-associated proteins by targeting peptides displayed on MHC receptors. However, cross-reactivities of these agents to off-target cells and tissues have resulted in serious, sometimes fatal, adverse events. We have developed a high-throughput genetic platform (termed "PresentER") that encodes MHC-I peptide minigenes for functional immunologic assays and determines the reactivities of TCR-like therapeutic agents against large libraries of MHC-I ligands. In this article, we demonstrated that PresentER could be used to identify the on-and-off targets of T cells and TCR-mimic (TCRm) antibodies using in vitro coculture assays or binding assays. We found dozens of MHC-I ligands that were cross-reactive with two TCRm antibodies and two native TCRs and that were not easily predictable by other methods.


Subject(s)
Cross Reactions/immunology , High-Throughput Screening Assays/methods , Histocompatibility Antigens Class I/genetics , Major Histocompatibility Complex/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Disease Models, Animal , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology
5.
Cancer Discov ; 10(3): 440-459, 2020 03.
Article in English | MEDLINE | ID: mdl-31915197

ABSTRACT

CREBBP mutations are highly recurrent in B-cell lymphomas and either inactivate its histone acetyltransferase (HAT) domain or truncate the protein. Herein, we show that these two classes of mutations yield different degrees of disruption of the epigenome, with HAT mutations being more severe and associated with inferior clinical outcome. Genes perturbed by CREBBP mutation are direct targets of the BCL6-HDAC3 onco-repressor complex. Accordingly, we show that HDAC3-selective inhibitors reverse CREBBP-mutant aberrant epigenetic programming, resulting in: (i) growth inhibition of lymphoma cells through induction of BCL6 target genes such as CDKN1A and (ii) restoration of immune surveillance due to induction of BCL6-repressed IFN pathway and antigen-presenting genes. By reactivating these genes, exposure to HDAC3 inhibitors restored the ability of tumor-infiltrating lymphocytes to kill DLBCL cells in an MHC class I and II-dependent manner, and synergized with PD-L1 blockade in a syngeneic model in vivo. Hence, HDAC3 inhibition represents a novel mechanism-based immune epigenetic therapy for CREBBP-mutant lymphomas. SIGNIFICANCE: We have leveraged the molecular characterization of different types of CREBBP mutations to define a rational approach for targeting these mutations through selective inhibition of HDAC3. This represents an attractive therapeutic avenue for targeting synthetic vulnerabilities in CREBBP-mutant cells in tandem with promoting antitumor immunity.This article is highlighted in the In This Issue feature, p. 327.


Subject(s)
CREB-Binding Protein/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Histone Deacetylases/genetics , Lymphoma/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , B7-H1 Antigen/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Epigenome/genetics , Epigenome/immunology , Genes, MHC Class I/immunology , Histocompatibility Antigens Class II/immunology , Histone Acetyltransferases/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/drug effects , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune System/drug effects , Immune System/immunology , Interferons/genetics , Interferons/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma/drug therapy , Lymphoma/immunology , Lymphoma/pathology , Mice , Mutation/genetics , Signal Transduction/drug effects
6.
Cancer Immunol Res ; 7(12): 1984-1997, 2019 12.
Article in English | MEDLINE | ID: mdl-31540894

ABSTRACT

T-cell immunotherapies are often thwarted by the limited presentation of tumor-specific antigens abetted by the downregulation of human leukocyte antigen (HLA). We showed that drugs inhibiting ALK and RET produced dose-related increases in cell-surface HLA in tumor cells bearing these mutated kinases in vitro and in vivo, as well as elevated transcript and protein expression of HLA and other antigen-processing machinery. Subsequent analysis of HLA-presented peptides after ALK and RET inhibitor treatment identified large changes in the immunopeptidome with the appearance of hundreds of new antigens, including T-cell epitopes associated with impaired peptide processing (TEIPP) peptides. ALK inhibition additionally decreased PD-L1 levels by 75%. Therefore, these oncogenes may enhance cancer formation by allowing tumors to evade the immune system by downregulating HLA expression. Altogether, RET and ALK inhibitors could enhance T-cell-based immunotherapies by upregulating HLA, decreasing checkpoint blockade ligands, and revealing new, immunogenic, cancer-associated antigens.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Antigens, Neoplasm/immunology , Histocompatibility Antigens Class I/immunology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Animals , Antigen Presentation/drug effects , Cell Line, Tumor , Crizotinib/pharmacology , Female , Humans , Mice, Transgenic , Neoplasms/immunology , Peptides/immunology , Pyrimidines/pharmacology , Sulfones/pharmacology
7.
Elife ; 72018 11 30.
Article in English | MEDLINE | ID: mdl-30499773

ABSTRACT

Tumors often co-exist with T cells that recognize somatically mutated peptides presented by cancer cells on major histocompatibility complex I (MHC-I). However, it is unknown why the immune system fails to eliminate immune-recognizable neoplasms before they manifest as frank disease. To understand the determinants of MHC-I peptide immunogenicity in nascent tumors, we tested the ability of thousands of MHC-I ligands to cause tumor subclone rejection in immunocompetent mice by use of a new 'PresentER' antigen presentation platform. Surprisingly, we show that immunogenic tumor antigens do not lead to immune-mediated cell rejection when the fraction of cells bearing each antigen ('clonal fraction') is low. Moreover, the clonal fraction necessary to lead to rejection of immunogenic tumor subclones depends on the antigen. These data indicate that tumor neoantigen heterogeneity has an underappreciated impact on immune elimination of cancer cells and has implications for the design of immunotherapeutics such as cancer vaccines.


Subject(s)
Clone Cells/pathology , Neoplasms/immunology , Neoplasms/pathology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Base Sequence , Bystander Effect , Cell Line, Tumor , Cytotoxicity, Immunologic , Gene Library , Immunocompetence , Major Histocompatibility Complex/immunology , Mice, Inbred C57BL , Peptides/immunology , Receptors for Activated C Kinase/immunology , T-Lymphocytes/immunology , Vaccination
9.
Immunity ; 48(4): 730-744.e5, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29669251

ABSTRACT

Although characterization of T cell exhaustion has unlocked powerful immunotherapies, the mechanisms sustaining adaptations of short-lived innate cells to chronic inflammatory settings remain unknown. During murine chronic viral infection, we found that concerted events in bone marrow and spleen mediated by type I interferon (IFN-I) and Toll-like receptor 7 (TLR7) maintained a pool of functionally exhausted plasmacytoid dendritic cells (pDCs). In the bone marrow, IFN-I compromised the number and the developmental capacity of pDC progenitors, which generated dysfunctional pDCs. Concurrently, exhausted pDCs in the periphery were maintained by self-renewal via IFN-I- and TLR7-induced proliferation of CD4- subsets. On the other hand, pDC functional loss was mediated by TLR7, leading to compromised IFN-I production and resistance to secondary infection. These findings unveil the mechanisms sustaining a self-perpetuating pool of functionally exhausted pDCs and provide a framework for deciphering long-term exhaustion of other short-lived innate cells during chronic inflammation.


Subject(s)
Cell Self Renewal/immunology , Dendritic Cells/immunology , Interferon Type I/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Membrane Glycoproteins/immunology , Toll-Like Receptor 7/immunology , 3T3 Cells , Animals , Carrier Proteins/biosynthesis , Cell Line , Cell Proliferation , DNA-Binding Proteins/biosynthesis , Dendritic Cells/cytology , Humans , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/biosynthesis , Repressor Proteins , Signal Transduction/immunology , Transcription Factor 4/biosynthesis , Transcription Factors/biosynthesis
11.
J Clin Invest ; 127(7): 2705-2718, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28628042

ABSTRACT

Preferentially expressed antigen in melanoma (PRAME) is a cancer-testis antigen that is expressed in many cancers and leukemias. In healthy tissue, PRAME expression is limited to the testes and ovaries, making it a highly attractive cancer target. PRAME is an intracellular protein that cannot currently be drugged. After proteasomal processing, the PRAME300-309 peptide ALYVDSLFFL (ALY) is presented in the context of human leukocyte antigen HLA-A*02:01 molecules for recognition by the T cell receptor (TCR) of cytotoxic T cells. Here, we have described Pr20, a TCR mimic (TCRm) human IgG1 antibody that recognizes the cell-surface ALY peptide/HLA-A2 complex. Pr20 is an immunological tool and potential therapeutic agent. Pr20 bound to PRAME+HLA-A2+ cancers. An afucosylated Fc form (Pr20M) directed antibody-dependent cellular cytotoxicity against PRAME+HLA-A2+ leukemia cells and was therapeutically effective against mouse xenograft models of human leukemia. In some tumors, Pr20 binding markedly increased upon IFN-γ treatment, mediated by induction of the immunoproteasome catalytic subunit ß5i. The immunoproteasome reduced internal destructive cleavages within the ALY epitope compared with the constitutive proteasome. The data provide rationale for developing TCRm antibodies as therapeutic agents for cancer, offer mechanistic insight on proteasomal regulation of tumor-associated peptide/HLA antigen complexes, and yield possible therapeutic solutions to target antigens with ultra-low surface presentation.


Subject(s)
Antigens, Neoplasm/immunology , HLA-A1 Antigen/immunology , Immunoglobulin G/pharmacology , Neoplasms, Experimental , Receptors, Antigen, T-Cell/immunology , Animals , Cell Line, Tumor , Female , Humans , Immunoglobulin G/immunology , Male , Mice , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Xenograft Model Antitumor Assays
12.
JCI Insight ; 2(6): e90196, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28352655

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) frequently harbors genetic alterations that activate the B cell receptor (BCR) and TLR pathways, which converge to activate NF-κB. While selective inhibition of BTK with ibrutinib causes clinical responses in relapsed DLBCL patients, most responses are partial and of a short duration. Here, we demonstrated that MyD88 silencing enhanced ibrutinib efficacy in DLBCL cells harboring MyD88 L265P mutations. Chemical downregulation of MyD88 expression with HDAC inhibitors also synergized with ibrutinib. We demonstrate that HDAC inhibitor regulation of MyD88 expression is mediated by STAT3. In turn, STAT3 silencing caused a decrease in MyD88 mRNA and protein levels, and enhanced the ibrutinib antilymphoma effect in MyD88 mutant DLBCL cells. Induced mutations in the STAT3 binding site in the MyD88 promotor region was associated with a decrease in MyD88 transcriptional activity. We also demonstrate that treatment with the HDAC inhibitor panobinostat decreased phosphorylated STAT3 binding to the MyD88 promotor. Accordingly, combined treatment with panobinostat and ibrutinib resulted in enhanced inhibition of NF-κB activity and caused regression of DLBCL xenografts. Our data provide a mechanistic rationale for combining HDAC inhibitors and ibrutinib for the treatment of DLBCL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mutation , Myeloid Differentiation Factor 88/genetics , Panobinostat/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Adenine/analogs & derivatives , Animals , Cell Line, Tumor , Drug Synergism , Humans , Mice , Piperidines , Promoter Regions, Genetic , STAT3 Transcription Factor/metabolism , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
13.
Cancer Immunol Res ; 4(11): 936-947, 2016 11.
Article in English | MEDLINE | ID: mdl-27680026

ABSTRACT

The major histocompatibility complex I (MHC-1) presents antigenic peptides to tumor-specific CD8+ T cells. The regulation of MHC-I by kinases is largely unstudied, even though many patients with cancer are receiving therapeutic kinase inhibitors. Regulators of cell-surface HLA amounts were discovered using a pooled human kinome shRNA interference-based approach. Hits scoring highly were subsequently validated by additional RNAi and pharmacologic inhibitors. MAP2K1 (MEK), EGFR, and RET were validated as negative regulators of MHC-I expression and antigen presentation machinery in multiple cancer types, acting through an ERK output-dependent mechanism; the pathways responsible for increased MHC-I upon kinase inhibition were mapped. Activated MAPK signaling in mouse tumors in vivo suppressed components of MHC-I and the antigen presentation machinery. Pharmacologic inhibition of MAPK signaling also led to improved peptide/MHC target recognition and killing by T cells and TCR-mimic antibodies. Druggable kinases may thus serve as immediately applicable targets for modulating immunotherapy for many diseases. Cancer Immunol Res; 4(11); 936-47. ©2016 AACR.


Subject(s)
Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/genetics , Neoplasms/genetics , Neoplasms/metabolism , Phosphotransferases/metabolism , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Disease Models, Animal , HLA-A Antigens/genetics , HLA-A Antigens/immunology , HLA-A Antigens/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Immunotherapy , MAP Kinase Signaling System , Melanoma, Experimental , Mice , Mice, Transgenic , Neoplasms/immunology , Programmed Cell Death 1 Receptor/metabolism , RNA Interference , RNA, Small Interfering/genetics
14.
Expert Opin Biol Ther ; 16(8): 979-87, 2016 08.
Article in English | MEDLINE | ID: mdl-27094818

ABSTRACT

INTRODUCTION: Monoclonal antibodies (mAbs) are potent cancer therapeutic agents, but exclusively recognize cell-surface targets whereas most cancer-associated proteins are found intracellularly. Hence, potential cancer therapy targets such as over expressed self-proteins, activated oncogenes, mutated tumor suppressors, and translocated gene products are not accessible to traditional mAb therapy. An emerging approach to target these epitopes is the use of TCR mimic mAbs (TCRm) that recognize epitopes similar to those of T cell receptors (TCR). AREAS COVERED: TCRm antigens are composed of a linear peptide sequence derived from degraded proteins and presented in the context of cell-surface MHC molecules. We discuss how the nature of the TCRm epitopes provides both advantages (absolute tumor specificity and access to a new universe of important targets) and disadvantages (low density, MHC restriction, MHC down-regulation, and cross-reactive linear epitopes) to conventional mAb therapy. We will also discuss potential solutions to these obstacles. EXPERT OPINION: TCRm combine the specificity of TCR recognition with the potency, pharmacologic properties, and versatility of mAbs. The structure and presentation of a TCRm epitope has important consequences related to the choice of targets, mAb design, available peptides and MHC subtype restrictions, possible cross-reactivity, and therapeutic activity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Animals , Antibodies, Neoplasm/immunology , Epitopes/immunology , Humans , Molecular Mimicry
15.
Oncoimmunology ; 5(1): e1049803, 2016.
Article in English | MEDLINE | ID: mdl-26942058

ABSTRACT

The major hurdle to the creation of cancer-specific monoclonal antibodies (mAb) exhibiting limited cross-reactivity with healthy human cells is the paucity of known tumor-specific or mutated protein epitopes expressed on the cancer cell surface. Mutated and overexpressed oncoproteins are typically cytoplasmic or nuclear. Cells can present peptides from these distinguishing proteins on their cell surface in the context of human leukocyte antigen (HLA). T cell receptor mimic (TCRm) mAb can be discovered that react specifically to these complexes, allowing for selective targeting of cancer cells. The state-of-the-art for TCRm and the challenges and opportunities are discussed. Several such TCRm are moving toward clinical trials now.

SELECTION OF CITATIONS
SEARCH DETAIL