Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 209
Filter
1.
J Biol Chem ; 300(4): 107153, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38462163

ABSTRACT

The innate immune system features a web of interacting pathways that require exquisite regulation. To identify novel nodes in this immune landscape, we conducted a gain-of-function, genome-wide CRISPR activation screen with influenza A virus. We identified both appreciated and novel antiviral genes, including Jade family PHD zinc finger 3 (JADE3) a protein involved in directing the histone acetyltransferase histone acetyltransferase binding to ORC1 complex to modify chromatin and regulate transcription. JADE3 is both necessary and sufficient to restrict influenza A virus infection. Our results suggest a distinct function for JADE3 as expression of the closely related paralogs JADE1 and JADE2 does not confer resistance to influenza A virus infection. JADE3 is required for both constitutive and inducible expression of the well-characterized antiviral gene interferon-induced transmembrane protein 3 (IFITM3). Furthermore, we find JADE3 activates the NF-kB signaling pathway, which is required for the promotion of IFITM3 expression by JADE3. Therefore, we propose JADE3 activates an antiviral genetic program involving NF-kB-dependent IFITM3 expression to restrict influenza A virus infection.


Subject(s)
Gene Expression Regulation , Immunity, Innate , Membrane Proteins , NF-kappa B , Oncogene Proteins , RNA-Binding Proteins , Animals , Humans , CRISPR-Cas Systems , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , HEK293 Cells , Immunity, Innate/genetics , Influenza A virus/immunology , Influenza, Human/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , NF-kappa B/genetics , NF-kappa B/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Signal Transduction , Oncogene Proteins/genetics , Oncogene Proteins/immunology
2.
Nature ; 623(7988): 820-827, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37938771

ABSTRACT

The majority of oncogenic drivers are intracellular proteins, constraining their immunotherapeutic targeting to mutated peptides (neoantigens) presented by individual human leukocyte antigen (HLA) allotypes1. However, most cancers have a modest mutational burden that is insufficient for generating responses using neoantigen-based therapies2,3. Neuroblastoma is a paediatric cancer that harbours few mutations and is instead driven by epigenetically deregulated transcriptional networks4. Here we show that the neuroblastoma immunopeptidome is enriched with peptides derived from proteins essential for tumorigenesis. We focused on targeting the unmutated peptide QYNPIRTTF discovered on HLA-A*24:02, which is derived from the neuroblastoma-dependency gene and master transcriptional regulator PHOX2B. To target QYNPIRTTF, we developed peptide-centric chimeric antigen receptors (PC-CARs) through a counter panning strategy using predicted potentially cross-reactive peptides. We further proposed that PC-CARs can recognize peptides on additional HLA allotypes when presenting a similar overall molecular surface. Informed by our computational modelling results, we show that PHOX2B PC-CARs also recognize QYNPIRTTF presented by HLA-A*23:01, the most common non-A2 allele in people with African ancestry. Finally, we demonstrate potent and specific killing of neuroblastoma cells expressing these HLAs in vitro and complete tumour regression in mice. These data suggest that PC-CARs have the potential to expand the pool of immunotherapeutic targets to include non-immunogenic intracellular oncoproteins and allow targeting through additional HLA allotypes in a clinical setting.


Subject(s)
Antigens, Neoplasm , Neuroblastoma , Oncogene Proteins , Peptides , Receptors, Chimeric Antigen , Animals , Humans , Mice , Africa/ethnology , Alleles , Amino Acid Sequence , Carcinogenesis , Cross Reactions , HLA-A Antigens/chemistry , HLA-A Antigens/immunology , Neuroblastoma/genetics , Neuroblastoma/immunology , Neuroblastoma/therapy , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/immunology , Peptides/antagonists & inhibitors , Peptides/chemistry , Peptides/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use
4.
Nature ; 599(7885): 477-484, 2021 11.
Article in English | MEDLINE | ID: mdl-34732890

ABSTRACT

The majority of oncogenic drivers are intracellular proteins, thus constraining their immunotherapeutic targeting to mutated peptides (neoantigens) presented by individual human leukocyte antigen (HLA) allotypes1. However, most cancers have a modest mutational burden that is insufficient to generate responses using neoantigen-based therapies2,3. Neuroblastoma is a paediatric cancer that harbours few mutations and is instead driven by epigenetically deregulated transcriptional networks4. Here we show that the neuroblastoma immunopeptidome is enriched with peptides derived from proteins that are essential for tumourigenesis and focus on targeting the unmutated peptide QYNPIRTTF, discovered on HLA-A*24:02, which is derived from the neuroblastoma dependency gene and master transcriptional regulator PHOX2B. To target QYNPIRTTF, we developed peptide-centric chimeric antigen receptors (CARs) using a counter-panning strategy with predicted potentially cross-reactive peptides. We further hypothesized that peptide-centric CARs could recognize peptides on additional HLA allotypes when presented in a similar manner. Informed by computational modelling, we showed that PHOX2B peptide-centric CARs also recognize QYNPIRTTF presented by HLA-A*23:01 and the highly divergent HLA-B*14:02. Finally, we demonstrated potent and specific killing of neuroblastoma cells expressing these HLAs in vitro and complete tumour regression in mice. These data suggest that peptide-centric CARs have the potential to vastly expand the pool of immunotherapeutic targets to include non-immunogenic intracellular oncoproteins and widen the population of patients who would benefit from such therapy by breaking conventional HLA restriction.


Subject(s)
Antigens, Neoplasm/immunology , HLA Antigens/immunology , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Oncogene Proteins/immunology , Receptors, Chimeric Antigen/immunology , Animals , Antigens, Neoplasm/metabolism , Cell Line , Cell Line, Tumor , Cross Reactions , Cross-Priming , Female , HLA Antigens/metabolism , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Humans , Interferon-gamma/immunology , Mice , Neoplasms/metabolism , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/metabolism , T-Lymphocytes/immunology , Transcription Factors/immunology , Transcription Factors/metabolism
5.
Biochim Biophys Acta Rev Cancer ; 1876(1): 188558, 2021 08.
Article in English | MEDLINE | ID: mdl-33933558

ABSTRACT

Increasing evidence shows that numerous cancer-testis antigens (CTAs) are uniquely overexpressed in various types of cancer and most CTAs are oncogenic. Overexpression of oncogenic CTAs promotes carcinogenesis, cancer metastasis, and drug resistance. Oncogenic CTAs are generally associated with poor prognosis in cancer patients and are an important hallmark of cancer, making them a crucial target for cancer immunotherapy. CTAs-targeted antibodies, vaccines, and chimeric antigen receptor-modified T cells (CAR-T) have recently been used in cancer treatment and achieved promising outcomes in the preclinical and early clinical trials. However, the efficacy of current CTA-targeted therapeutics is either moderate or low in cancer therapy. CTA-targeted cancer immunotherapy is facing enormous challenges. Several critical scientific problems need to be resolved: (1) the antigen presentation function of MHC-I protein is usually deficient in cancer patients, so that very low amounts of intracellular CTA epitopes are presented to tumor cell membrane surface, leading to weak immune response and subsequent immunity to CTAs; (2) various immunosuppressive cells are rich in tumor tissues leading to diminished tumor immunity; (3) the tumor tissue microenvironment markedly reduces the efficacy of cancer immunotherapy. In the current review paper, the authors propose new strategies and approaches to overcome the barriers of CTAs-targeted immunotherapy and to develop novel potent immune therapeutics against cancer. Finally, we highlight that the oncogenic CTAs have high tumor specificity and immunogenicity, and are sensible targets for cancer immunotherapy. We predict that CTAs-targeted immunotherapy will bring about breakthroughs in cancer therapy in the near future.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents, Immunological/therapeutic use , Cancer Vaccines/therapeutic use , Neoplasms/therapy , Oncogene Proteins/metabolism , T-Lymphocytes/transplantation , Testis/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/adverse effects , Cancer Vaccines/adverse effects , Humans , Immunotherapy, Adoptive/adverse effects , Male , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Testis/immunology , Treatment Outcome , Tumor Microenvironment
6.
Leukemia ; 34(6): 1626-1636, 2020 06.
Article in English | MEDLINE | ID: mdl-31908357

ABSTRACT

Immunotherapy targeting leukemia-associated antigens has shown promising results. Because of the heterogeneity of leukemia, vaccines with a single peptide have elicited only a limited immune response. Targeting several peptides together elicited peptide-specific cytotoxic T lymphocytes (CTLs) in leukemia patients, and this was associated with clinical responses. Thus, the discovery of novel antigens is essential. In the current study, we investigated cyclin E as a novel target for immunotherapy. Cyclin E1 and cyclin E2 were found to be highly expressed in hematologic malignancies, according to reverse transcription polymerase chain reaction and western blot analysis. We identified two HLA-A*0201 binding nonameric peptides, CCNE1M from cyclin E1 and CCNE2L from cyclin E2, which both elicited the peptide-specific CTLs. The peptide-specific CTLs specifically kill leukemia cells. Furthermore, CCNE1M and CCNE2L CTLs were increased in leukemia patients who underwent allogeneic hematopoietic stem cell transplantation, and this was associated with desired clinical outcomes. Our findings suggest that cyclin E1 and cyclin E2 are potential targets for immunotherapy in leukemia.


Subject(s)
Antigens, Neoplasm/metabolism , Cancer Vaccines/immunology , Cyclin E/immunology , Cyclins/immunology , HLA-A2 Antigen/immunology , Leukemia/immunology , Oncogene Proteins/immunology , Adult , Aged , Antigens, Neoplasm/immunology , Cancer Vaccines/pharmacology , Epitopes, T-Lymphocyte/immunology , Female , Humans , Male , Middle Aged , T-Lymphocytes, Cytotoxic/immunology , Young Adult
7.
Cell Signal ; 66: 109443, 2020 02.
Article in English | MEDLINE | ID: mdl-31626955

ABSTRACT

The ability of phagocytes to recognize, immobilize, and engulf extracellular targets are fundamental immune cell processes that allow for the destruction of a variety of microbial intruders. The phagocytic process depends onsignalling events that initiate dynamic changes in the plasma membrane architecture that are required to accommodate the internalization of large particulate targets. To better understand fundamental molecular mechanisms responsible for facilitating phagocytic receptor-mediated regulation of cytoskeletal networks, our research has focused on investigating representative immunoregulatory proteins from the channel catfish (Ictalurus punctatus) leukocyte immune-type receptor family (IpLITRs). Specifically, we have shown that a specific IpLITR-type can regulate the constitutive deployment of filopodial-like structures to actively capture and secure targets to the phagocyte surface, which is followed by F-actin mediated membrane dynamics that are associated with the formation of phagocytic cup-like structures that precede target engulfment. In the present study, we use confocal imaging to examine the recruitment of mediators of the F-actin cytoskeleton during IpLITR-mediated regulation of membrane dynamics. Our results provide novel details regarding the dynamic recruitment of the signaling effectors Nck and Syk during classical as well as atypical IpLITR-induced phagocytic processes.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Ictaluridae/immunology , Oncogene Proteins/immunology , Phagocytosis/immunology , Receptors, Immunologic/immunology , Syk Kinase/immunology , Animals , Cell Line , Fibroblasts , Pseudopodia/immunology , Rats
8.
Radiographics ; 39(7): 2069-2084, 2019.
Article in English | MEDLINE | ID: mdl-31697628

ABSTRACT

Merkel cell carcinoma (MCC) is a rare and aggressive cutaneous neuroendocrine tumor with a higher mortality rate than melanoma. Approximately 40% of MCC patients have nodal or distant metastasis at initial presentation, and one-third of patients will develop distant metastatic disease over their clinical course. Although MCC is rare, its incidence has been steadily increasing. Furthermore, the immunogenicity of MCC and its diagnostic and therapeutic application have made MCC one of the most rapidly developing topics in dermatology and oncology. Owing to the aggressive and complex nature of MCC, a multidisciplinary approach is necessary for management of this tumor, including dermatologists, surgeons, radiation oncologists, medical oncologists, pathologists, radiologists, and nuclear medicine physicians. Imaging plays a crucial role in diagnosis, planning for surgery or radiation therapy, and assessment of treatment response and surveillance. However, MCC is still not well recognized among radiologists and nuclear medicine physicians, likely owing to its rarity. The purpose of this review is to raise awareness of MCC among imaging experts by describing the epidemiology, pathophysiology, and clinical features of MCC and current clinical management with a focus on the role of imaging. The authors highlight imaging findings characteristic of MCC, as well as the clinical significance of CT, MRI, sentinel lymph node mapping, fluorine 18 fluorodeoxyglucose PET/CT, and other nuclear medicine studies such as bone scintigraphy and somatostatin receptor scintigraphy. ©RSNA, 2019.


Subject(s)
Carcinoma, Merkel Cell/diagnostic imaging , Magnetic Resonance Imaging/methods , Positron Emission Tomography Computed Tomography/methods , Skin Neoplasms/diagnostic imaging , Antibodies, Viral/blood , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/secondary , Carcinoma, Merkel Cell/secondary , Carcinoma, Merkel Cell/virology , Humans , Lymphatic Metastasis/diagnostic imaging , Merkel cell polyomavirus/isolation & purification , Neoplasm Staging , Oncogene Proteins/immunology , Polyomavirus Infections/diagnostic imaging , Polyomavirus Infections/virology , Prognosis , Radiopharmaceuticals/analysis , Radiopharmaceuticals/pharmacokinetics , Receptors, Somatostatin/drug effects , Sentinel Lymph Node Biopsy , Skin Neoplasms/virology , Tumor Virus Infections/diagnostic imaging , Tumor Virus Infections/virology , Viral Proteins/immunology
9.
Cell Rep ; 29(4): 889-903.e10, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31644911

ABSTRACT

Notwithstanding the positive clinical impact of endocrine therapies in estrogen receptor-alpha (ERα)-positive breast cancer, de novo and acquired resistance limits the therapeutic lifespan of existing drugs. Taking the position that resistance is nearly inevitable, we undertook a study to identify and exploit targetable vulnerabilities that were manifest in endocrine therapy-resistant disease. Using cellular and mouse models of endocrine therapy-sensitive and endocrine therapy-resistant breast cancer, together with contemporary discovery platforms, we identified a targetable pathway that is composed of the transcription factors FOXA1 and GRHL2, a coregulated target gene, the membrane receptor LYPD3, and the LYPD3 ligand, AGR2. Inhibition of the activity of this pathway using blocking antibodies directed against LYPD3 or AGR2 inhibits the growth of endocrine therapy-resistant tumors in mice, providing the rationale for near-term clinical development of humanized antibodies directed against these proteins.


Subject(s)
Hepatocyte Nuclear Factor 3-alpha/metabolism , Mammary Neoplasms, Experimental/metabolism , Transcription Factors/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor alpha/genetics , Female , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mice , Mucoproteins/immunology , Mucoproteins/metabolism , Oncogene Proteins/immunology , Oncogene Proteins/metabolism
10.
BMC Cancer ; 19(1): 670, 2019 Jul 08.
Article in English | MEDLINE | ID: mdl-31286874

ABSTRACT

BACKGROUND: In epithelial cells, tyrosine kinases induce tyrosine phosphorylation and ubiquitination of the E-cadherin complex, which is responsible for the epithelial-mesenchymal transition (EMT). However, the precise mechanisms remain unclear. METHODS: Protein antibody microarray analysis and E3 ligase profiling were performed to detect the unique E3 ligase underlying E-cadherin downregulation in lung adenocarcinoma tissues. Gene knockdown was performed using viral shRNA. Immunoblotting, immunofluorescence, immunoprecipitation, and xenograft models in vivo were integratively applied to explore RNF43-induced EMT in lung adenocarcinoma cell lines. RESULTS: Protein antibody microarray analysis and E3 ligase profiling revealed that the RING finger protein 43 (RNF43) was linked to E-cadherin downregulation within the context of c-Src activation in lung adenocarcinoma tissues. In addition, the c-Src-Caspase-8 interaction markedly increased c-Src activity. Activated c-Src phosphorylated E-cadherin at the tyrosine 797 site to initiate RNF43-mediated E-cadherin ubiquitination at lysine 816 and subsequent degradation, thus allowing the nuclear translocation of ß-catenin and upregulation of Vimentin and RNF43 expression in lung adenocarcinoma cells. Decreased E-cadherin expression and increased Vimentin expression induced the EMT phenotype and promoted tumor metastasis. The Frizzled 8 (Frz8)-RNF43-induced ubiquitination of phosphorylated E-cadherin was blocked by a monoclonal antibody against the cysteine-rich domain (CRD) of Frz8 but not by antibodies against the protease domain (PA) of RNF43. CONCLUSIONS: Our data suggest that RNF43 participates in the regulation of EMT in the metastasis of lung adenocarcinoma through the ubiquitination and degradation of phosphorylated E-cadherin by activated c-Src.


Subject(s)
Adenocarcinoma of Lung/secondary , Antigens, CD/metabolism , CSK Tyrosine-Protein Kinase/metabolism , Cadherins/metabolism , DNA-Binding Proteins/metabolism , Epithelial-Mesenchymal Transition , Lung Neoplasms/pathology , Oncogene Proteins/metabolism , A549 Cells , Adenocarcinoma of Lung/metabolism , Animals , Antibodies, Monoclonal , Caspase 8/metabolism , Cells, Cultured , DNA-Binding Proteins/immunology , Follow-Up Studies , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Oncogene Proteins/immunology , Phosphorylation , Proteolysis , Receptors, Cell Surface/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Vimentin/metabolism , beta Catenin/metabolism
11.
Nat Commun ; 10(1): 2935, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31270335

ABSTRACT

Trace elements play important roles in human health, but little is known about their functions in humoral immunity. Here, we show an important role for iron in inducing cyclin E and B cell proliferation. We find that iron-deficient individuals exhibit a significantly reduced antibody response to the measles vaccine when compared to iron-normal controls. Mice with iron deficiency also exhibit attenuated T-dependent or T-independent antigen-specific antibody responses. We show that iron is essential for B cell proliferation; both iron deficiency and α-ketoglutarate inhibition could suppress cyclin E1 induction and S phase entry of B cells upon activation. Finally, we demonstrate that three demethylases, KDM2B, KDM3B and KDM4C, are responsible for histone 3 lysine 9 (H3K9) demethylation at the cyclin E1 promoter, cyclin E1 induction and B cell proliferation. Thus, our data reveal a crucial role of H3K9 demethylation in B cell proliferation, and the importance of iron in humoral immunity.


Subject(s)
B-Lymphocytes/immunology , Cell Proliferation , Histones/chemistry , Histones/immunology , Immunity, Humoral , Lysine/immunology , Animals , B-Lymphocytes/chemistry , B-Lymphocytes/cytology , Cell Cycle , Cells, Cultured , Cyclin E/genetics , Cyclin E/immunology , Demethylation , F-Box Proteins/genetics , F-Box Proteins/immunology , Histones/genetics , Iron/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/immunology , Lymphocyte Activation , Lysine/genetics , Mice , Mice, Inbred C57BL , Oncogene Proteins/genetics , Oncogene Proteins/immunology , Promoter Regions, Genetic , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
J Leukoc Biol ; 106(5): 1101-1115, 2019 11.
Article in English | MEDLINE | ID: mdl-31216372

ABSTRACT

Neutrophils play a central role in immunity and inflammation via their intrinsic ability to migrate into inflamed tissue, to phagocytose pathogens, and to kill bacterial and fungi by releasing large quantities of superoxide anions and lytic enzymes. The molecular pathways controlling neutrophil microbicidal functions are still unclear, because neutrophils have a short half-life and are resistant to genetic manipulation. Neutrophil-like cells (NLC) can be generated from myeloid progenitors conditionally immortalized with the ER-HoxB8 oncoprotein, but whether these cells can replace neutrophils in high-throughput functional assays is unclear. Here, we assess the ability of NLC derived from ER-HoxB8 progenitors to produce ROS and to perform chemotaxis and phagocytosis. We compare the Ca2+ responses and effector functions of NLC to primary murine neutrophils and document the molecular basis of their functional differences by mRNA profiling. Pro-inflammatory cytokines enhanced the expression by NLC of neutrophil surface markers and transcription factors. Ca2+ elevations evoked in NLC by agonists, adhesion receptors, and store depletion resembled the physiological responses recorded in primary neutrophils, but NLC expressed reduced amounts of Ca2+ signaling proteins and of chemotactic receptors. Unlike their myeloid progenitors, NLC produced H2 O2 when adhered to fibronectin, migrated toward chemotactic peptides, phagocytosed opsonized particles, and generated intracellular ROS. NLC phagocytosed as efficiently as primary neutrophils but produced 50 times less ROS and migrated less efficiently toward chemoattractant. Our data indicate that NLC can replace neutrophils to study Ca2+ signaling and phagocytosis, but that their incomplete granulocytic differentiation limits their use for chemotaxis and ROS production assays.


Subject(s)
Bone Marrow Cells/immunology , Gene Expression Regulation/immunology , Homeodomain Proteins/immunology , Neutrophils/immunology , Oncogene Proteins/immunology , Signal Transduction/immunology , Animals , Bone Marrow Cells/cytology , Calcium Signaling/genetics , Calcium Signaling/immunology , Homeodomain Proteins/genetics , Male , Mice , Mice, Transgenic , Neutrophils/cytology , Oncogene Proteins/genetics , Reactive Oxygen Species/immunology , Signal Transduction/genetics
13.
Nat Med ; 25(5): 767-775, 2019 05.
Article in English | MEDLINE | ID: mdl-31011208

ABSTRACT

Anti-tumor immunity is driven by self versus non-self discrimination. Many immunotherapeutic approaches to cancer have taken advantage of tumor neoantigens derived from somatic mutations. Here, we demonstrate that gene fusions are a source of immunogenic neoantigens that can mediate responses to immunotherapy. We identified an exceptional responder with metastatic head and neck cancer who experienced a complete response to immune checkpoint inhibitor therapy, despite a low mutational load and minimal pre-treatment immune infiltration in the tumor. Using whole-genome sequencing and RNA sequencing, we identified a novel gene fusion and demonstrated that it produces a neoantigen that can specifically elicit a host cytotoxic T cell response. In a cohort of head and neck tumors with low mutation burden, minimal immune infiltration and prevalent gene fusions, we also identified gene fusion-derived neoantigens that generate cytotoxic T cell responses. Finally, analyzing additional datasets of fusion-positive cancers, including checkpoint-inhibitor-treated tumors, we found evidence of immune surveillance resulting in negative selective pressure against gene fusion-derived neoantigens. These findings highlight an important class of tumor-specific antigens and have implications for targeting gene fusion events in cancers that would otherwise be less poised for response to immunotherapy, including cancers with low mutational load and minimal immune infiltration.


Subject(s)
Antigens, Neoplasm/genetics , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/immunology , Gene Fusion , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Humans , NFI Transcription Factors/genetics , NFI Transcription Factors/immunology , Neoplasms/genetics , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Oncogene Proteins/genetics , Oncogene Proteins/immunology , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/therapy , Whole Genome Sequencing
14.
Front Immunol ; 10: 415, 2019.
Article in English | MEDLINE | ID: mdl-30930895

ABSTRACT

Growing evidence indicate that large antigen-containing particles induce potent T cell-dependent high-affinity antibody responses. These responses require large particle internalization after recognition by the B cell receptor (BCR) on B cells. However, the molecular mechanisms governing BCR-mediated internalization remain unclear. Here we use a high-throughput quantitative image analysis approach to discriminate between B cell particle binding and internalization. We systematically show, using small molecule inhibitors, that human B cells require a SYK-dependent IgM-BCR signaling transduction via PI3K to efficiently internalize large anti-IgM-coated particles. IgM-BCR-mediated activation of PI3K involves both the adaptor protein NCK and the co-receptor CD19. Interestingly, we here reveal a strong NCK-dependence without profound requirement of the co-receptor CD19 in B cell responses to large particles. Furthermore, we demonstrate that the IgM-BCR/NCK signaling event facilitates RAC1 activation to promote actin cytoskeleton remodeling necessary for particle engulfment. Thus, we establish NCK/PI3K/RAC1 as an attractive IgM-BCR signaling axis for biological intervention to prevent undesired antibody responses to large particles.


Subject(s)
Antigen Presentation/immunology , B-Lymphocytes/immunology , Lymphocyte Activation/immunology , Phagocytosis/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/immunology , B-Lymphocytes/metabolism , Humans , Immunoglobulin M/immunology , Oncogene Proteins/immunology , Phosphatidylinositol 3-Kinases/immunology , Receptors, Antigen, B-Cell/immunology , rac1 GTP-Binding Protein/immunology
15.
PLoS One ; 13(6): e0199197, 2018.
Article in English | MEDLINE | ID: mdl-29958295

ABSTRACT

The activity of Cullin-RING ubiquitin E3 ligases (CRL) is regulated by NEDD8 modification. DCN-like proteins promote Cullin neddylation as scaffold-like E3s. One DCNL, DCNL5, is highly expressed in immune tissue. Here, we provide evidence that DCNL5 may be involved in innate immunity, as it is a direct substrate of the kinase IKKα during immune signalling. We find that upon activation of Toll-like receptors, DCNL5 gets rapidly and transiently phosphorylated on a specific N-terminal serine residue (S41). This phosphorylation event is specifically mediated by IKKα and not IKKß. Our data for the first time provides evidence that DCNL proteins are post-translationally modified in an inducible manner. Our findings also provide the first example of a DCNL member as a kinase substrate in a signalling pathway, indicating that the activity of at least some DCNLs may be regulated.


Subject(s)
I-kappa B Kinase/immunology , Immunity, Innate , Oncogene Proteins/immunology , Peptide Synthases/immunology , Signal Transduction/immunology , Animals , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Mice , NEDD8 Protein/genetics , NEDD8 Protein/immunology , Oncogene Proteins/genetics , Peptide Synthases/genetics , Phosphorylation/genetics , Phosphorylation/immunology , RAW 264.7 Cells , Signal Transduction/genetics
16.
JCI Insight ; 3(12)2018 06 21.
Article in English | MEDLINE | ID: mdl-29925694

ABSTRACT

Mucin 1 (MUC1) is a heterodimeric protein that is aberrantly overexpressed on the surface of diverse human carcinomas and is an attractive target for the development of mAb-based therapeutics. However, attempts at targeting the shed MUC1 N-terminal subunit have been unsuccessful. We report here the generation of mAb 3D1 against the nonshed oncogenic MUC1 C-terminal (MUC1-C) subunit. We show that mAb 3D1 binds with low nM affinity to the MUC1-C extracellular domain at the restricted α3 helix. mAb 3D1 reactivity is selective for MUC1-C-expressing human cancer cell lines and primary cancer cells. Internalization of mAb 3D1 into cancer cells further supported the conjugation of mAb 3D1 to monomethyl auristatin E (MMAE). The mAb 3D1-MMAE antibody-drug conjugate (ADC) (a) kills MUC1-C-positive cells in vitro, (b) is nontoxic in MUC1-transgenic (MUC1.Tg) mice, and (c) is active against human HCC827 lung tumor xenografts. Humanized mAb (humAb) 3D1 conjugated to MMAE also exhibited antitumor activity in (a) MUC1.Tg mice harboring syngeneic MC-38/MUC1 tumors, (b) nude mice bearing human ZR-75-1 breast tumors, and (c) NCG mice engrafted with a patient-derived triple-negative breast cancer. These findings and the absence of associated toxicities support clinical development of humAb 3D1-MMAE ADCs as a therapeutic for the many cancers with MUC1-C overexpression.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunoconjugates/pharmacology , Mucin-1/immunology , Oncogene Proteins/immunology , Animals , Antibodies, Monoclonal/chemistry , Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Carcinoma/pathology , Cell Line, Tumor , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Models, Molecular , Mucin-1/chemistry , Oligopeptides , Oncogene Proteins/chemistry , Protein Conformation, alpha-Helical , Xenograft Model Antitumor Assays
17.
Oncol Rep ; 39(5): 2385-2392, 2018 May.
Article in English | MEDLINE | ID: mdl-29498403

ABSTRACT

Cancer vaccines have been developed as a new therapeutic approach, however, their clinical benefit remains limited. We previously performed a phase II study for advanced colorectal cancer (CRC) using five human leukocyte antigen (HLA-A*24:02)-restricted peptides derived from kinase of the outer chloroplast membrane 1, translocase of outer mitochondrial membrane 34 (TOMM34), ring finger protein 43 (RNF43), vascular endothelial growth factor receptor 1 (VEGFR1) and VEGFR2. In the present study the relationship between overall survival (OS) and several biomarkers, including cytotoxic T lymphocyte (CTL) and immunoglobulin G (IgG) responses to these five peptides, was investigated. In 89 advanced CRC patients treated with a combination therapy consisting of these five peptides and oxaliplatin-based chemotherapy, plasma was collected before and after 3 months of vaccine administration. IgGs reactive to each of the five peptides were assessed using the multiplex bead suspension Luminex system. Antigen-specific T-cell responses were estimated by enzyme-linked immunoSpot assay. Plasma levels of TOMM34 IgG (P<0.001), RNF43 IgG (P<0.001) and VEGFR2 IgG (P<0.001) were significantly increased after vaccination and stronger VEGFR2 IgG responses correlated significantly with OS in HLA-matched patients (P=0.034). CTL responses to VEGFR1 and VEGFR2 were also significantly increased in the HLA-matched group (P=0.049 and P<0.001, respectively). However, increased CTL response did not correlate with OS. Multivariate analysis indicated that IgG responses to VEGFR2 were the most significant predictor for OS in the HLA-A*24:02-matched group (P=0.04). Our findings indicated that VEGFR2 IgG responses may be an important immunological biomarker in the early course of treatment for CRC patients treated with therapeutic epitope peptides.


Subject(s)
Cancer Vaccines/administration & dosage , Colorectal Neoplasms/drug therapy , HLA-A24 Antigen/immunology , Immunoglobulin G/metabolism , Aged , Cancer Vaccines/immunology , Colorectal Neoplasms/immunology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/immunology , Double-Blind Method , Epitopes/immunology , Female , Humans , Male , Middle Aged , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/immunology , Mitochondrial Precursor Protein Import Complex Proteins , Oncogene Proteins/chemistry , Oncogene Proteins/immunology , Survival Analysis , Treatment Outcome , Ubiquitin-Protein Ligases , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-1/immunology , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/immunology
18.
Cancer Sci ; 109(5): 1545-1551, 2018 May.
Article in English | MEDLINE | ID: mdl-29473265

ABSTRACT

We previously reported a phase I clinical trial of a peptide vaccine ring finger protein 43 (RNF43) and 34-kDa translocase of the outer mitochondrial membrane (TOMM34) combined with uracil-tegafur (UFT)/LV for patients with metastatic colorectal cancer (CRC), and demonstrated the safety and immunological responsiveness of this combination therapy. In this study, we evaluated vaccination-induced immune responses to clarify the survival benefit of the combination therapy as adjuvant treatment. We enrolled 44 patients initially in an HLA-masked fashion. After the disclosure of HLA, 28 patients were in the HLA-A*2402-matched and 16 were in the unmatched group. In the HLA-matched group, 14 patients had positive CTL responses specific for the RNF43 and/or TOMM34 peptides after 2 cycles of treatment and 9 had negative responses; in the HLA-unmatched group, 10 CTL responses were positive and 2 negative. In the HLA-matched group, 3-year relapse-free survival (RFS) was significantly better in the positive CTL subgroup than in the negative-response subgroup. Patients with negative vaccination-induced CTL responses showed a significant trend towards shorter RFS than those with positive responses. Moreover, in the HLA-unmatched group, the positive CTL response subgroup showed an equally good 3-year RFS as in the HLA-matched group. In conclusion, vaccination-induced CTL response to peptide vaccination could predict survival in the adjuvant setting for stage III CRC.


Subject(s)
Colorectal Neoplasms/mortality , DNA-Binding Proteins/immunology , Mitochondrial Membrane Transport Proteins/immunology , Oncogene Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccination , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Double-Blind Method , Female , HLA-A24 Antigen/immunology , Humans , Male , Middle Aged , Mitochondrial Precursor Protein Import Complex Proteins , Neoplasm Staging , Ubiquitin-Protein Ligases
19.
Arthritis Rheumatol ; 70(4): 594-605, 2018 04.
Article in English | MEDLINE | ID: mdl-29287303

ABSTRACT

OBJECTIVE: The nuclear oncoprotein DEK is an autoantigen associated with juvenile idiopathic arthritis (JIA), especially the oligoarticular subtype. DEK is a secreted chemotactic factor. Abundant levels of DEK and DEK autoantibodies are found in inflamed synovium in JIA. We undertook this study to further characterize the nature of DEK autoantibodies in screening serum samples from 2 different cohorts that consisted mostly of patients with JIA. METHODS: DEK autoantibody levels were analyzed in sera from 33 JIA patients, 13 patients with other inflammatory conditions, and 11 healthy controls, as well as in 89 serum samples from JIA patients receiving anti-tumor necrosis factor (anti-TNF) therapy. Recombinant His-tagged full-length DEK protein (1-375 amino acids [aa]) and the 187-375-aa and 1-350-aa His-tagged DEK fragments made in a baculovirus system were used for enzyme-linked immunosorbent assay (ELISA) and immunoblotting. The C-terminal 25-aa fragment of DEK was expressed in a glutathione S-transferase-tagged vector. ELISA results were calculated as area under the curve by the trapezoidal rule. RESULTS: DEK autoantibody levels were significantly higher in patients with polyarticular JIA than in those with oligoarticular JIA, and were higher in patients with polyarticular JIA who had more active disease after cessation of anti-TNF therapy. Immunoblotting against the C-terminal 25-aa fragment of DEK confirmed that this section of the DEK molecule is the most immunogenic domain. CONCLUSION: DEK autoantibody levels are higher in patients with polyarticular JIA than in those with oligoarticular JIA, and higher in patients who have disease flares after cessation of anti-TNF therapy. The C-terminal 25-aa fragment is the most immunogenic portion of DEK. These findings are significant with respect to the nature of DEK autoantibodies, their contribution to JIA pathogenesis, and their implications for JIA management.


Subject(s)
Antirheumatic Agents/immunology , Arthritis, Juvenile/blood , Autoantibodies/blood , Chromosomal Proteins, Non-Histone/immunology , Oncogene Proteins/immunology , Poly-ADP-Ribose Binding Proteins/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adolescent , Arthritis, Juvenile/drug therapy , Arthritis, Juvenile/immunology , Autoantibodies/immunology , Case-Control Studies , Child , Female , Humans , Male , Symptom Flare Up , Withholding Treatment
20.
Sci Signal ; 10(498)2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28951536

ABSTRACT

There is intense interest in developing therapeutic strategies for RAS proteins, the most frequently mutated oncoprotein family in cancer. Development of effective anti-RAS therapies will be aided by the greater appreciation of RAS isoform-specific differences in signaling events that support neoplastic cell growth. However, critical issues that require resolution to facilitate the success of these efforts remain. In particular, the use of well-validated anti-RAS antibodies is essential for accurate interpretation of experimental data. We evaluated 22 commercially available anti-RAS antibodies with a set of distinct reagents and cell lines for their specificity and selectivity in recognizing the intended RAS isoforms and mutants. Reliability varied substantially. For example, we found that some pan- or isoform-selective anti-RAS antibodies did not adequately recognize their intended target or showed greater selectivity for another; some were valid for detecting G12D and G12V mutant RAS proteins in Western blotting, but none were valid for immunofluorescence or immunohistochemical analyses; and some antibodies recognized nonspecific bands in lysates from "Rasless" cells expressing the oncoprotein BRAFV600E Using our validated antibodies, we identified RAS isoform-specific siRNAs and shRNAs. Our results may help to ensure the accurate interpretation of future RAS studies.


Subject(s)
Antineoplastic Agents, Immunological/immunology , Mutation , Oncogene Proteins/immunology , ras Proteins/immunology , Animals , Antineoplastic Agents, Immunological/analysis , Cell Line, Tumor , Fibroblasts , Humans , Hybridomas , Mice , Oncogene Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/immunology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , RNA, Small Interfering , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...