Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Int J Cancer ; 147(8): 2293-2302, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32468570

ABSTRACT

Immune checkpoint inhibition leads to response in some patients with head and neck squamous cell carcinoma (HNSCC). Robust biomarkers are lacking to date. We analyzed viral status, gene expression signatures, mutational load and mutational signatures in whole exome and RNA-sequencing data of the HNSCC TCGA dataset (n = 496) and a validation set (DKTK MASTER cohort, n = 10). Public single-cell gene expression data from 17 HPV-negative HNSCC were separately reanalyzed. APOBEC3-associated TCW motif mutations but not total single nucleotide variant burden were significantly associated with inflammation. This association was restricted to HPV-negative HNSCC samples. An APOBEC-enriched, HPV-negative subgroup was identified, that showed higher T-cell inflammation and immune checkpoint expression, as well as expression of APOBEC3 genes. Mutations in immune-evasion pathways were also enriched in these tumors. Analysis of single-cell sequencing data identified expression of APOBEC3B and 3C genes in malignant cells. We identified an APOBEC-enriched subgroup of HPV-negative HNSCC with a distinct immunogenic phenotype, potentially mediating response to immunotherapy.


Subject(s)
APOBEC Deaminases/genetics , APOBEC Deaminases/immunology , Head and Neck Neoplasms/immunology , Immune Evasion/genetics , Immune Evasion/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Cohort Studies , Exome/genetics , Exome/immunology , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Head and Neck Neoplasms/virology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/virology , Male , Middle Aged , Mutation/genetics , Papillomaviridae/immunology , Papillomavirus Infections/genetics , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Sequence Analysis, RNA/methods , Squamous Cell Carcinoma of Head and Neck/virology , T-Lymphocytes/immunology , Transcriptome/genetics , Transcriptome/immunology
2.
Pharmacol Res ; 153: 104683, 2020 03.
Article in English | MEDLINE | ID: mdl-32050092

ABSTRACT

Triple negative breast cancer (TNBC) is most aggressive subtype of breast cancers with high probability of metastasis as well as lack of specific targets and targeted therapeutics. TNBC is characterized with unique tumor microenvironment (TME), which differs from other subtypes. TME is associated with induction of proliferation, angiogenesis, inhibition of apoptosis and immune system suppression, and drug resistance. Exosomes are promising nanovesicles, which orchestrate the TME by communicating with different cells within TME. The components of TME including transformed ECM, soluble factors, immune suppressive cells, epigenetic modifications and re-programmed fibroblasts together hamper antitumor response and helps progression and metastasis of TNBCs. Therefore, TME could be a therapeutic target of TNBC. The current review presents latest updates on the role of exosomes in modulation of TME, approaches for targeting TME and combination of immune checkpoint inhibitors and target chemotherapeutics. Finally, we also discussed various phytochemicals that alter genetic, transcriptomic and proteomic profiles of TME along with current challenges and future implications. Thus, as TME is associated with the hallmarks of TNBC, the understanding of the impact of different components can improve the clinical benefits of TNBC patients.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Exome/drug effects , Triple Negative Breast Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Animals , Apoptosis/drug effects , Epigenesis, Genetic , Exome/immunology , Female , Humans , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
3.
Biomed Res Int ; 2019: 2183510, 2019.
Article in English | MEDLINE | ID: mdl-31781598

ABSTRACT

Immunotherapy directed against cancer-specific neoantigens derived from non-silent mutants is a promising individualized strategy for cancer treatment. Neoantigens shared across patients could be used as a public resource for developing T cell-based therapy. To identify potential public neoantigens for therapy in gastric cancer (GC), 74 GC patients were enrolled in this study. Combined with the TCGA cohort and other published studies, whole exome sequencing data from 942 GC patients were used to detect somatic mutations and predict neoantigens shared by GC patients. The mutations pattern between our study and the TCGA cohort is comparable, and C > T is the most common substitution. The number of neoantigens was significantly higher in older patients (age ≥60) compared to younger patients (age <60), both in this study and the TCGA cohort. Recurrent neoantigens were found in eight genes (TP53, PIK3CA, PGM5, ERBB3, C6, TRIM49C, OR4C16, and KRAS) in this study. The neoantigen-associated mutations PIK3CA (p.H1047R) and TP53 (p.R175H) are common across several cancer types, indicating their potential usage. Overall, our study illustrates a comprehensive genomic landscape of GC and provides the recurrent neoantigens to facilitate further immunotherapy.


Subject(s)
Antigens, Neoplasm/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Stomach Neoplasms/immunology , Tumor Suppressor Protein p53/genetics , Aged , Antigens, Neoplasm/immunology , Cell- and Tissue-Based Therapy/methods , Class I Phosphatidylinositol 3-Kinases/immunology , Databases, Genetic , Exome/genetics , Exome/immunology , Female , Genome, Human/genetics , Genome, Human/immunology , Genomics , Humans , Immunotherapy/methods , Male , Mutation/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy , T-Lymphocytes/immunology , Tumor Suppressor Protein p53/immunology
4.
Biomed Res Int ; 2019: 8103142, 2019.
Article in English | MEDLINE | ID: mdl-31312661

ABSTRACT

Neoantigens are optimal tumor-specific targets for T-cell based immunotherapy, especially for patients with "undruggable" mutated driver genes. T-cell immunotherapy can be a "universal" treatment for HLA genotype patients sharing same oncogenic mutations. To identify potential neoantigens for therapy in gastric cancer, 32 gastric cancer patients were enrolled in our study. Whole exome sequencing data from these patients was processed by TSNAD software to detect cancer somatic mutations and predict neoantigens. The somatic mutations between different patients suggested a high interpatient heterogeneity. C>A and C>T substitutions are common, suggesting an active nucleotide excision repair. The number of predicted neoantigens was significantly higher in patients at stage T1a compared to in patients at T2 or T4b. Six genes (PIK3CA, FAT4, BRCA2, GNAQ, LRP1B, and PREX2) were found as recurrently mutated driver genes in our study. Combining with highly frequent HLA alleles, several neoantigens derived from six recurrently mutated genes were considered as potential targets for further immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Precision Medicine , Stomach Neoplasms/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/genetics , Antigens, Neoplasm/isolation & purification , Antigens, Neoplasm/therapeutic use , Databases, Genetic , Exome/genetics , Exome/immunology , Female , High-Throughput Nucleotide Sequencing , Humans , Immunologic Factors/genetics , Immunologic Factors/therapeutic use , Immunotherapy , Male , Middle Aged , Mutation , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy
5.
Br J Cancer ; 120(9): 922-930, 2019 04.
Article in English | MEDLINE | ID: mdl-30894686

ABSTRACT

BACKGROUND: Approximately 4% of colorectal cancer (CRC) patients have at least two simultaneous cancers in the colon. Due to the shared environment, these synchronous CRCs (SCRCs) provide a unique setting to study colorectal carcinogenesis. Understanding whether these tumours are genetically similar or distinct is essential when designing therapeutic approaches. METHODS: We performed exome sequencing of 47 primary cancers and corresponding normal samples from 23 patients. Additionally, we carried out a comprehensive mutational signature analysis to assess whether tumours had undergone similar mutational processes and the first immune cell score analysis (IS) of SCRC to analyse the interplay between immune cell invasion and mutation profile in both lesions of an individual. RESULTS: The tumour pairs shared only few mutations, favouring different mutations in known CRC genes and signalling pathways and displayed variation in their signature content. Two tumour pairs had discordant mismatch repair statuses. In majority of the pairs, IS varied between primaries. Differences were not explained by any clinicopathological variable or mutation burden. CONCLUSIONS: The study shows major diversity within SCRCs. Rather than rely on data from one tumour, our study highlights the need to evaluate both tumours of a synchronous pair for optimised targeted therapy.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Lymphocytes/immunology , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/immunology , Aged , Aged, 80 and over , CD3 Complex/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Case-Control Studies , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Exome/genetics , Exome/immunology , Female , Humans , Lymphocytes/pathology , Male , Microsatellite Instability , Middle Aged , Mutation , Neoplasms, Multiple Primary/pathology
6.
Genet Med ; 21(1): 243-251, 2019 01.
Article in English | MEDLINE | ID: mdl-29921932

ABSTRACT

PURPOSE: The etiology of 80% of patients with primary antibody deficiency (PAD), the second most common type of human immune system disorder after human immunodeficiency virus infection, is yet unknown. METHODS: Clinical/immunological phenotyping and exome sequencing of a cohort of 126 PAD patients (55.5% male, 95.2% childhood onset) born to predominantly consanguineous parents (82.5%) with unknown genetic defects were performed. The American College of Medical Genetics and Genomics criteria were used for validation of pathogenicity of the variants. RESULTS: This genetic approach and subsequent immunological investigations identified potential disease-causing variants in 86 patients (68.2%); however, 27 of these patients (31.4%) carried autosomal dominant (24.4%) and X-linked (7%) gene defects. This genetic approach led to the identification of new phenotypes in 19 known genes (38 patients) and the discovery of a new genetic defect (CD70 pathogenic variants in 2 patients). Medical implications of a definite genetic diagnosis were reported in ~50% of the patients. CONCLUSION: Due to misclassification of the conventional approach for targeted sequencing, employing next-generation sequencing as a preliminary step of molecular diagnostic approach to patients with PAD is crucial for management and treatment of the patients and their family members.


Subject(s)
Genetic Predisposition to Disease , Genetic Testing , Genomics , Immune System Diseases/genetics , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Exome/genetics , Exome/immunology , Female , High-Throughput Nucleotide Sequencing , Humans , Immune System Diseases/immunology , Immune System Diseases/pathology , Male , Mutation , Phenotype , Sequence Analysis, DNA , Exome Sequencing , Young Adult
7.
Breast Cancer Res Treat ; 173(1): 167-177, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30229447

ABSTRACT

PURPOSE: Immune characterizations of cancers, including breast cancer, have led to information useful for prognoses and are considered to be important in the future of refining the use of immunotherapies, including immune checkpoint inhibitor therapies. In this study, we sought to extend these characterizations with genomics approaches, particularly with cost-effective employment of exome files. METHODS: By recovery of immune receptor recombination reads from the cancer genome atlas (TCGA) breast cancer dataset, we observed associations of these recombinations with T-cell and B-cell biomarkers and with distinct survival rates. RESULTS: Recovery of TRD or IGH recombination reads was associated with an improved disease-free survival (p = 0.047 and 0.045, respectively). Determination of the HLA types using the exome files allowed matching of T-cell receptor V- and J-gene segment usage with specific HLA alleles, in turn allowing a refinement of the association of immune receptor recombination read recoveries with survival. For example, the TRBV7, HLA-C*07:01 combination represented a significantly worse, disease-free outcome (p = 0.014) compared to all other breast cancer samples. By direct comparisons of distinct TRB gene segment usage, HLA allele combinations revealed breast cancer subgroups, within the entire TCGA breast cancer dataset with even more dramatic survival distinctions. CONCLUSIONS: In sum, the use of exome files for recovery of adaptive immune receptor recombination reads, and the simultaneous determination of HLA types, has the potential of advancing the use of immunogenomics for immune characterization of breast tumor samples.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/mortality , HLA Antigens/genetics , Receptors, Antigen, T-Cell/genetics , Recombination, Genetic , Disease-Free Survival , Exome/genetics , Exome/immunology , Female , HLA Antigens/immunology , Humans , Immunoglobulin Heavy Chains/genetics , Receptors, Antigen, T-Cell/immunology , Survival Rate
8.
Methods Mol Biol ; 1884: 203-214, 2019.
Article in English | MEDLINE | ID: mdl-30465205

ABSTRACT

Neo-antigens expressed on tumors are targets for development of cancer immunotherapy strategies. Use of prediction algorithms to identify neo-antigens yields a significant number of peptides that must be validated in laborious and time-consuming methods; many prove to be false-positive identifications. The use of HLA peptidomics allows the isolation of the HLA-peptide complexes directly from cells and can be done on fresh tumor, patient-derived xerographs, or cell lines when the tissue sample is limited. This method can be used to identify both HLA class I and HLA class II or any different MHC from different species. Here we describe the steps to create the immune-affinity columns used from the process, the immunoprecipitation procedure, and also the isolation of the peptides that will be analyzed by mass spectrometry.


Subject(s)
Antigens, Neoplasm/isolation & purification , Exome/immunology , Neoplasms/immunology , Proteomics/methods , Tandem Mass Spectrometry/methods , Algorithms , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Chromatography, High Pressure Liquid/instrumentation , Chromatography, High Pressure Liquid/methods , Exome/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/isolation & purification , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/isolation & purification , Humans , Hybridomas , Immunoprecipitation/instrumentation , Immunoprecipitation/methods , Neoplasms/pathology , Proteomics/instrumentation , Spectrometry, Mass, Electrospray Ionization/instrumentation , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/instrumentation
11.
Oncotarget ; 7(5): 5110-7, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26819371

ABSTRACT

The antigenicity of cells is demarcated by the peptides bound by their Human Leucocyte Antigen (HLA) molecules. Through this antigen presentation, T cell specificity response is controlled. As a fraction of the expressed mutated peptides is presented on the HLA, these neo-epitopes could be immunogenic. Such neo-antigens have recently been identified through screening for predicted mutated peptides, using synthetic peptides or ones expressed from minigenes, combined with screening of patient tumor-infiltrating lymphocytes (TILs). Here we present a time and cost-effective method that combines whole-exome sequencing analysis with HLA peptidome mass spectrometry, to identify neo-antigens in a melanoma patient. Of the 1,019 amino acid changes identified through exome sequencing, two were confirmed by mass spectrometry to be presented by the cells. We then synthesized peptides and evaluated the two mutated neo-antigens for reactivity with autologous bulk TILs, and found that one yielded mutant-specific T-cell response. Our results demonstrate that this method can be used for immune response prediction and promise to provide an alternative approach for identifying immunogenic neo-epitopes in cancer.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Exome/immunology , Histocompatibility Antigens Class I/immunology , Peptidomimetics/immunology , Humans
12.
Trends Immunol ; 37(2): 126-140, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26791050

ABSTRACT

Recent advances in genome analysis have provided important insights into the genetic architecture of infectious and inflammatory diseases. The combined analysis of loci detected by genome-wide association studies (GWAS) in 22 inflammatory diseases has revealed a shared genetic core and associated biochemical pathways that play a central role in pathological inflammation. Parallel whole-exome sequencing studies have identified 265 genes mutated in primary immunodeficiencies (PID). Here, we examine the overlap between these two data sets, and find that it consists of genes essential for protection against infections and in which persistent activation causes pathological inflammation. Based on this intersection, we propose that, although strong or inactivating mutations (rare variants) in these genes may cause severe disease (PIDs), their more subtle modulation potentially by common regulatory/coding variants may contribute to chronic inflammation.


Subject(s)
Autoimmune Diseases/genetics , Genetic Loci/immunology , Immunologic Deficiency Syndromes/genetics , Animals , Exome/immunology , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Mutation/genetics , Risk
13.
Cancer Immunol Res ; 3(9): 992-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26048577

ABSTRACT

The ability to use circulating peripheral blood cells and matched tumor sequencing data as a basis for neoantigen prediction has exciting possibilities for application in the personalized treatment of cancer patients. We have used a high-throughput screening approach, combining whole-exome sequence data, mRNA microarrays, and publicly available epitope prediction algorithm output to identify mutated proteins processed and displayed by patient tumors and recognized by circulating immune cells. Matched autologous melanoma cell lines and peripheral blood mononuclear cells were used to create mixed lymphocyte tumor cell cultures, resulting in an expansion of tumor-reactive T cells to use for mutated peptide screening. Five patients were investigated, three of whom had a durable complete response (CR; 15+ years) in an autologous melanoma-pulsed dendritic cell clinical trial. We identified seven mutated antigens in total that stimulated T-effector memory cells in two of the five patients. While the procedure did not result in clinically applicable neoantigens for all patients, those identified were likely important in tumor clearance, leading to durable CR. The nature of the screening process allows results to be obtained rapidly and is easily applicable to a wide variety of different tumor types.


Subject(s)
Antigens, Neoplasm/genetics , Exome/immunology , Melanoma/genetics , Melanoma/immunology , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Exome/genetics , Humans , Interferon-gamma/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Mutation
14.
Inflamm Bowel Dis ; 21(6): 1229-36, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25895113

ABSTRACT

BACKGROUND: Pediatric Inflammatory Bowel Disease (PIBD) is a chronic condition seen in genetically predisposed individuals. Genome-wide association studies have implicated >160 genomic loci in IBD with many genes coding for proteins in key immune pathways. This study looks at autoimmune disease burden in patients diagnosed with PIBD and interrogates exome data of a subset of patients. METHODS: Patients were recruited from the Southampton Genetics of PIBD cohort. Clinical diagnosis of autoimmune disease in these individuals was ascertained from medical records. For a subset of patients with PIBD and concurrent asthma, exome data was interrogated to ascertain the burden of pathogenic variants within genes implicated in asthma. Association testing was conducted between cases and population controls using the SKAT-O test. RESULTS: Forty-nine (28.3%) PIBD children (18.49% CD, 8.6% UC, and 21.15% IBDU patients) had a concurrent clinical diagnosis of at least one other autoimmune disorder; asthma was the most prevalent, affecting 16.2% of the PIBD cohort. Rare and common variant association testing revealed 6 significant genes (P < 0.05) before Bonferroni adjustment. Three of these genes were previously implicated in both asthma and IBD (ZPBP2 IL1R1, and IL18R1) and 3 in asthma only (PYHIN1, IL2RB, and GSTP1). CONCLUSIONS: One-third of our cohort had a concurrent autoimmune condition. We observed higher incidence of asthma compared with the overall pediatric prevalence. Despite a small sample size, SKAT-O evaluated a significant burden of rare and common mutations in 6 genes. Variant burden suggests that a systemic immune dysregulation rather than organ-specific could underpin immune dysfunction for a subset of patients.


Subject(s)
Autoimmune Diseases/genetics , Colitis, Ulcerative/genetics , Crohn Disease/genetics , Exome , Adolescent , Asthma/genetics , Autoimmune Diseases/epidemiology , Child , Cohort Studies , Colitis, Ulcerative/epidemiology , Comorbidity , Crohn Disease/epidemiology , Egg Proteins/genetics , Egg Proteins/immunology , Exome/immunology , Female , Genetic Predisposition to Disease , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/immunology , Humans , Incidence , Interleukin-18 Receptor alpha Subunit/genetics , Interleukin-18 Receptor alpha Subunit/immunology , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/immunology
15.
J Clin Immunol ; 33(8): 1289-92, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24122031

ABSTRACT

We describe a boy who developed autoinflammatory (chronic sterile multifocal osteomyelitis) and autoimmune (autoimmune cytopenias; vitiligo) phenotypes who subsequently developed disseminated granulomatous disease. Whole exome sequencing revealed homozygous RAG1 mutations thus expanding the spectrum of combined immunodeficiency with autoimmunity and granuloma that can occur with RAG deficiency.


Subject(s)
Autoantibodies/biosynthesis , Exome/genetics , Exome/immunology , Granulomatous Disease, Chronic/immunology , Homeodomain Proteins/genetics , Immunologic Deficiency Syndromes/immunology , Mutation/immunology , Osteomyelitis/immunology , Child, Preschool , Granulomatous Disease, Chronic/genetics , Homozygote , Humans , Immunologic Deficiency Syndromes/genetics , Male , Osteomyelitis/genetics , Sequence Analysis, DNA
16.
PLoS One ; 7(9): e38670, 2012.
Article in English | MEDLINE | ID: mdl-23049726

ABSTRACT

Several studies have shown that cancers actively regulate alternative splicing. Altered splicing mechanisms in cancer lead to cancer-specific transcripts different from the pool of transcripts occurring only in healthy tissue. At the same time, altered presentation of HLA class I epitopes is frequently observed in various types of cancer. Down-regulation of genes related to HLA class I antigen processing has been observed in several cancer types, leading to fewer HLA class I antigens on the cell surface. Here, we use a peptidome wide analysis of predicted alternative splice forms, based on a publicly available database, to show that peptides over-represented in cancer splice variants comprise significantly fewer predicted HLA class I epitopes compared to peptides from normal transcripts. Peptides over-represented in cancer transcripts are in the case of the three most common HLA class I supertype representatives consistently found to contain fewer predicted epitopes compared to normal tissue. We observed a significant difference in amino acid composition between protein sequences associated with normal versus cancer tissue, as transcripts found in cancer are enriched with hydrophilic amino acids. This variation contributes to the observed significant lower likelihood of cancer-specific peptides to be predicted epitopes compared to peptides found in normal tissue.


Subject(s)
Alternative Splicing/immunology , Epitopes/genetics , Exome/genetics , HLA Antigens/genetics , Neoplasms/genetics , Peptides/genetics , RNA, Messenger/genetics , Alleles , Amino Acid Motifs , Databases, Genetic , Epitopes/immunology , Exome/immunology , Gene Frequency , HLA Antigens/immunology , Humans , Neoplasms/diagnosis , Neoplasms/immunology , Neoplasms/pathology , Peptides/immunology , Proteome/genetics , Proteome/immunology , RNA, Messenger/immunology
17.
J Allergy Clin Immunol ; 130(2): 481-8.e2, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22721650

ABSTRACT

BACKGROUND: Clinical immunology has traditionally relied on accurate phenotyping of the patient's immune dysfunction for the identification of a candidate gene or genes for sequencing and molecular confirmation. Although this is also true for other branches of medicine, the marked variability in immune-related phenotypes and the highly complex network of molecules that confer normal host immunity are challenges that clinical immunologists often face in their quest to establish a specific genetic diagnosis. OBJECTIVE: We sought to identify the underlying genetic cause in a consanguineous family with chronic inflammatory bowel disease-like disorder and combined immunodeficiency. METHODS: We performed exome sequencing followed by autozygome filtration. RESULTS: A truncating mutation in LPS-responsive beige-like anchor (LRBA), which abolished protein expression, was identified as the most likely candidate variant in this family. CONCLUSION: The combined exome sequencing and autozygosity mapping approach is a powerful tool in the study of atypical immune dysfunctions. We identify LRBA as a novel immunodeficiency candidate gene the precise role of which in the immune system requires future studies.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Exome/genetics , Inflammatory Bowel Diseases/genetics , Mutation , Severe Combined Immunodeficiency/genetics , Adaptor Proteins, Signal Transducing/deficiency , Adolescent , Base Sequence , Child , Consanguinity , DNA Mutational Analysis , Exome/immunology , Family , Humans , Immunophenotyping , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/immunology , Lipopolysaccharides/immunology , Male , Molecular Sequence Data , Pedigree , Phenotype , Severe Combined Immunodeficiency/complications , Severe Combined Immunodeficiency/immunology , Young Adult
19.
Nature ; 482(7385): 400-4, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22318521

ABSTRACT

Cancer immunoediting, the process by which the immune system controls tumour outgrowth and shapes tumour immunogenicity, is comprised of three phases: elimination, equilibrium and escape. Although many immune components that participate in this process are known, its underlying mechanisms remain poorly defined. A central tenet of cancer immunoediting is that T-cell recognition of tumour antigens drives the immunological destruction or sculpting of a developing cancer. However, our current understanding of tumour antigens comes largely from analyses of cancers that develop in immunocompetent hosts and thus may have already been edited. Little is known about the antigens expressed in nascent tumour cells, whether they are sufficient to induce protective antitumour immune responses or whether their expression is modulated by the immune system. Here, using massively parallel sequencing, we characterize expressed mutations in highly immunogenic methylcholanthrene-induced sarcomas derived from immunodeficient Rag2(-/-) mice that phenotypically resemble nascent primary tumour cells. Using class I prediction algorithms, we identify mutant spectrin-ß2 as a potential rejection antigen of the d42m1 sarcoma and validate this prediction by conventional antigen expression cloning and detection. We also demonstrate that cancer immunoediting of d42m1 occurs via a T-cell-dependent immunoselection process that promotes outgrowth of pre-existing tumour cell clones lacking highly antigenic mutant spectrin-ß2 and other potential strong antigens. These results demonstrate that the strong immunogenicity of an unedited tumour can be ascribed to expression of highly antigenic mutant proteins and show that outgrowth of tumour cells that lack these strong antigens via a T-cell-dependent immunoselection process represents one mechanism of cancer immunoediting.


Subject(s)
Exome/genetics , Exome/immunology , Immunologic Surveillance/immunology , Neoplasms/genetics , Neoplasms/immunology , T-Lymphocytes/immunology , Algorithms , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Histocompatibility Antigens Class I/immunology , Humans , Male , Methylcholanthrene , Mice , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Models, Immunological , Neoplasms/chemically induced , Neoplasms/pathology , Reproducibility of Results , Sarcoma/chemically induced , Sarcoma/genetics , Sarcoma/immunology , Sarcoma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...