Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
BMC Cancer ; 22(1): 2, 2022 Jan 03.
Article in English | MEDLINE | ID: mdl-34980012

ABSTRACT

BACKGROUND: Oncogenic metabolic reprogramming contributes to tumor growth and immune evasion. The intertumoral metabolic heterogeneity and interaction of distinct metabolic pathways may determine patient outcomes. In this study, we aim to determine the clinical and immunological significance of metabolic subtypes according to the expression levels of genes related to glycolysis and cholesterol-synthesis in bladder cancer (BCa). METHODS: Based on the median expression levels of glycolytic and cholesterogenic genes, patients were stratified into 4 subtypes (mixed, cholesterogenic, glycolytic, and quiescent) in an integrated cohort including TCGA, GSE13507, and IMvigor210. Clinical, genomic, transcriptomic, and tumor microenvironment characteristics were compared between the 4 subtypes. RESULTS: The 4 metabolic subtypes exhibited distinct clinical, molecular, and genomic patterns. Compared to quiescent subtype, mixed subtype was more likely to be basal tumors and was significantly associated with poorer prognosis even after controlling for age, gender, histological grade, clinical stage, and molecular phenotypes. Additionally, mixed tumors harbored a higher frequency of RB1 and LRP1B copy number deletion compared to quiescent tumors (25.7% vs. 12.7 and 27.9% vs. 10.2%, respectively, both adjusted P value< 0.05). Furthermore, aberrant PIK3CA expression level was significantly correlated with those of glycolytic and cholesterogenic genes. The quiescent subtype was associated with lower stemness indices and lower signature scores for gene sets involved in genomic instability, including DNA replication, DNA damage repair, mismatch repair, and homologous recombination genes. Moreover, quiescent tumors exhibited lower expression levels of pyruvate dehydrogenase kinases 1-3 (PDK1-3) than the other subtypes. In addition, distinct immune cell infiltration patterns were observed across the 4 metabolic subtypes, with greater infiltration of M0/M2 macrophages observed in glycolytic and mixed subtypes. However, no significant difference in immunotherapy response was observed across the 4 metabolic subtypes. CONCLUSION: This study proposed a new metabolic subtyping method for BCa based on genes involved in glycolysis and cholesterol synthesis pathways. Our findings may provide novel insight for the development of personalized subtype-specific treatment strategies targeting metabolic vulnerabilities.


Subject(s)
Cholesterol/biosynthesis , Glycolysis/genetics , Immune System/cytology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Class I Phosphatidylinositol 3-Kinases/genetics , DNA Copy Number Variations , DNA Repair/genetics , Databases, Genetic , Genomic Instability/genetics , Glycolysis/immunology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Oncogenes/genetics , Oncogenes/immunology , Polymorphism, Single Nucleotide , Prognosis , Receptors, LDL/genetics , Retinoblastoma Binding Proteins/genetics , Signal Transduction , Transcriptome , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Ubiquitin-Protein Ligases/genetics , Urinary Bladder Neoplasms/mortality
2.
PLoS Pathog ; 17(4): e1009141, 2021 04.
Article in English | MEDLINE | ID: mdl-33826675

ABSTRACT

HIV persists during antiretroviral therapy (ART) as integrated proviruses in cells descended from a small fraction of the CD4+ T cells infected prior to the initiation of ART. To better understand what controls HIV persistence and the distribution of integration sites (IS), we compared about 15,000 and 54,000 IS from individuals pre-ART and on ART, respectively, with approximately 395,000 IS from PBMC infected in vitro. The distribution of IS in vivo is quite similar to the distribution in PBMC, but modified by selection against proviruses in expressed genes, by selection for proviruses integrated into one of 7 specific genes, and by clonal expansion. Clones in which a provirus integrated in an oncogene contributed to cell survival comprised only a small fraction of the clones persisting in on ART. Mechanisms that do not involve the provirus, or its location in the host genome, are more important in determining which clones expand and persist.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/virology , Leukocytes, Mononuclear/virology , Oncogenes/genetics , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , DNA, Viral/genetics , Humans , Oncogenes/immunology , Proviruses/genetics , Virus Replication/genetics
3.
Front Immunol ; 12: 728853, 2021.
Article in English | MEDLINE | ID: mdl-35140701

ABSTRACT

Immunoglobulin M (IgM) autoantibodies, as the early appearing antibodies in humoral immunity when stimulated by antigens, might be excellent biomarkers for the early detection of lung cancer (LC). We aimed to develop a multi-analyte integrative model combining IgM autoantibodies and a traditional tumor biomarker that could be a valuable and powerful auxiliary diagnostic tool and might improve the accuracy of early detection of lung adenocarcinoma (LUAD). A customized protein array based on cancer driver genes was constructed and applied in the discovery cohort consisting of 68 LUAD patients and 68 normal controls (NCs); 31 differentially expressed IgM autoantibodies were identified. The top 5 candidate IgM autoantibodies [based on the area under the receiver operating characteristic curve (AUC) ranking], namely, TSHR, ERBB2, survivin, PIK3CA, and JAK2, were validated in the validation cohort using enzyme-linked immunosorbent assay (ELISA), which included 147 LUAD samples, 72 lung squamous cell carcinoma (LUSC) samples, 44 small cell lung carcinoma (SCLC) samples, and 147 NCs. These indicators presented diagnostic capacity for LUAD, with AUCs of 0.599, 0.613, 0.579, 0.601, and 0.633, respectively (p < 0.05). However, none of them showed a significant difference between the SCLC and NC groups, and only the IgM autoantibody against JAK2 showed a higher expression in LUSC than in NC (p = 0.046). Through logistic regression analysis, with the five IgM autoantibodies and carcinoembryonic antigen (CEA), one diagnostic model was constructed for LUAD. The model yielded an AUC of 0.827 (sensitivity = 56.63%, specificity = 93.98%). The diagnostic efficiency was superior to that of either CEA (AUC = 0.692) or IgM autoantibodies alone (AUC = 0.698). Notably, the accuracy of this model in early-stage LUAD reached 83.02%. In conclusion, we discovered and identified five novel IgM indicators and developed a multi-analyte model combining IgM autoantibodies and CEA, which could be a valuable and powerful auxiliary diagnostic tool and might improve the accuracy of early detection of LUAD.


Subject(s)
Adenocarcinoma of Lung/immunology , Autoantibodies/immunology , Carcinoembryonic Antigen/immunology , Immunoglobulin M/immunology , Lung Neoplasms/immunology , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/immunology , Biomarkers, Tumor/immunology , Carcinoma, Squamous Cell/immunology , Early Detection of Cancer/methods , Female , Humans , Male , Middle Aged , Oncogenes/immunology , ROC Curve
4.
Int J Mol Sci ; 23(1)2021 Dec 22.
Article in English | MEDLINE | ID: mdl-35008514

ABSTRACT

High ERß/HER oncogenic signaling defines lung tumors with an aggressive biology. We previously showed that combining the anti-estrogen fulvestrant with the pan-HER inhibitor dacomitinib reduced ER/HER crosstalk and produced synergistic anti-tumor effects in immunocompromised lung cancer models, including KRAS mutant adenocarcinoma. How this combination affects the tumor microenvironment (TME) is not known. We evaluated the effects of fulvestrant and dacomitinib on murine bone marrow-derived macrophages (BMDMs) and CD8+ T cells, and tested the efficacy of the combination in vivo, using the KRAS mutant syngeneic lung adenocarcinoma model, FVBW-17. While this combination synergistically inhibited proliferation of FVBW-17 cells, it had unwanted effects on immune cells, by reducing CD8+ T cell activity and phagocytosis in BMDMs and inducing PD-1. The effects were largely attributed to dacomitinib, which caused downregulation of Src family kinases and Syk in immune cells. In a subcutaneous flank model, the combination induced an inflamed TME with increased myeloid cells and CD8+ T cells and enhanced PD-1 expression in the splenic compartment. Concomitant administration of anti-PD-1 antibody with fulvestrant and dacomitinib was more efficacious than fulvestrant plus dacomitinib alone. Administering anti-PD-1 sequentially after fulvestrant plus dacomitinib was synergistic, with a two-fold greater tumor inhibitory effect compared to concomitant therapy, in both the flank model and in a lung metastasis model. Sequential triple therapy has potential for treating lung cancer that shows limited response to current therapies, such as KRAS mutant lung adenocarcinoma.


Subject(s)
Estrogen Receptor beta/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, ErbB-2/genetics , Tumor Microenvironment/genetics , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Cell Line, Tumor , Estrogen Receptor beta/immunology , Female , Humans , Immunotherapy/methods , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Oncogenes/genetics , Oncogenes/immunology , Programmed Cell Death 1 Receptor/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Quinazolinones/pharmacology , Receptor, ErbB-2/immunology , Tumor Microenvironment/immunology
5.
Cell Rep ; 33(8): 108426, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33238122

ABSTRACT

Gene expression is controlled by the collective binding of transcription factors to cis-regulatory regions. Deciphering gene-centered regulatory networks is vital to understanding and controlling gene misexpression in human disease; however, systematic approaches to uncovering regulatory networks have been lacking. Here we present high-throughput interrogation of gene-centered activation networks (HIGAN), a pipeline that employs a suite of multifaceted genomic approaches to connect upstream signaling inputs, trans-acting TFs, and cis-regulatory elements. We apply HIGAN to understand the aberrant activation of the cytidine deaminase APOBEC3B, an intrinsic source of cancer hypermutation. We reveal that nuclear factor κB (NF-κB) and AP-1 pathways are the most salient trans-acting inputs, with minor roles for other inflammatory pathways. We identify a cis-regulatory architecture dominated by a major intronic enhancer that requires coordinated NF-κB and AP-1 activity with secondary inputs from distal regulatory regions. Our data demonstrate how integration of cis and trans genomic screening platforms provides a paradigm for building gene-centered regulatory networks.


Subject(s)
Gene Expression/genetics , Gene Regulatory Networks/genetics , Oncogenes/immunology , Humans , Signal Transduction
6.
J Immunother Cancer ; 8(1)2020 04.
Article in English | MEDLINE | ID: mdl-32376723

ABSTRACT

BACKGROUND: An immune active cancer phenotype typified by a T helper 1 (Th-1) immune response has been associated with increased responsiveness to immunotherapy and favorable prognosis in some but not all cancer types. The reason of this differential prognostic connotation remains unknown. METHODS: To explore the contextual prognostic value of cancer immune phenotypes, we applied a multimodal pan-cancer analysis among 31 different histologies (9282 patients), encompassing immune and oncogenic transcriptomic analysis, mutational and neoantigen load and copy number variations. RESULTS: We demonstrated that the favorable prognostic connotation conferred by the presence of a Th-1 immune response was abolished in tumors displaying specific tumor-cell intrinsic attributes such as high transforming growth factor-beta (TGF-ß) signaling and low proliferation capacity. This observation was independent of mutation rate. We validated this observation in the context of immune checkpoint inhibition. WNT-ß catenin, barrier molecules, Notch, hedgehog, mismatch repair, telomerase activity and AMPK signaling were the pathways most coherently associated with an immune silent phenotype together with mutations of driver genes including IDH1/2, FOXA2, HDAC3, PSIP1, MAP3K1, KRAS, NRAS, EGFR, FGFR3, WNT5A and IRF7. CONCLUSIONS: This is the first systematic study demonstrating that the prognostic and predictive role of a bona fide favorable intratumoral immune response is dependent on the disposition of specific oncogenic pathways. This information could be used to refine stratification algorithms and prioritize hierarchically relevant targets for combination therapies.


Subject(s)
Gene Expression Profiling/methods , Immunity/immunology , Immunotherapy/methods , Neoplasms/immunology , Oncogenes/immunology , Female , Humans , Male , Neoplasms/mortality , Prognosis , Survival Analysis
7.
Nat Commun ; 11(1): 1000, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32081859

ABSTRACT

Long noncoding RNAs (lncRNAs) are emerging as critical regulators of gene expression and they play fundamental roles in immune regulation. Here we introduce an integrated algorithm, ImmLnc, for identifying lncRNA regulators of immune-related pathways. We comprehensively chart the landscape of lncRNA regulation in the immunome across 33 cancer types and show that cancers with similar tissue origin are likely to share lncRNA immune regulators. Moreover, the immune-related lncRNAs are likely to show expression perturbation in cancer and are significantly correlated with immune cell infiltration. ImmLnc can help prioritize cancer-related lncRNAs and further identify three molecular subtypes (proliferative, intermediate, and immunological) of non-small cell lung cancer. These subtypes are characterized by differences in mutation burden, immune cell infiltration, expression of immunomodulatory genes, response to chemotherapy, and prognosis. In summary, the ImmLnc pipeline and the resulting data serve as a valuable resource for understanding lncRNA function and to advance identification of immunotherapy targets.


Subject(s)
Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Neoplasms/genetics , Neoplasms/immunology , Oncogenes/immunology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology , Algorithms , Carcinoma, Non-Small-Cell Lung/classification , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Databases, Genetic , Humans , Lung Neoplasms/classification , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Neoplasms/classification
8.
Cancer Discov ; 10(4): 498-505, 2020 04.
Article in English | MEDLINE | ID: mdl-32094155

ABSTRACT

RET alterations have been characterized as oncogenic drivers in multiple cancers. The clinical validation of highly selective RET inhibitors demonstrates the utility of specific targeting of aberrantly activated RET in patients with cancers such as medullary thyroid cancer or non-small cell lung cancer. The remarkable responses observed have opened the field of RET-targeted inhibitors. In this review, we seek to focus on the impact of therapeutic RET targeting in cancers. SIGNIFICANCE: Successful clinical translation of selective RET inhibitors is poised to alter the therapeutic landscape of altered cancers. Questions that clearly need to be addressed relate to the ability to maintain long-term inhibition of tumor cell growth, how to prepare for the potential mechanisms of acquired resistance, and the development of next-generation selective RET inhibitors.


Subject(s)
Neoplasms/genetics , Oncogenes/immunology , Proto-Oncogene Proteins c-ret/immunology , Cell Line, Tumor , Humans
9.
Mol Oncol ; 14(1): 54-68, 2020 01.
Article in English | MEDLINE | ID: mdl-31736230

ABSTRACT

cMet is a well-characterized oncogene that is the target of many drugs including small molecule and biologic pathway inhibitors, and, more recently, antibody-drug conjugates (ADCs). However, the clinical benefit from cMet-targeted therapy has been limited. We developed a novel cMet-targeted 'third-generation' ADC, TR1801-ADC, that was optimized at different levels including specificity, stability, toxin-linker, conjugation site, and in vivo efficacy. Our nonagonistic cMet antibody was site-specifically conjugated to the pyrrolobenzodiazepine (PBD) toxin-linker tesirine and has picomolar activity in cancer cell lines derived from different solid tumors including lung, colorectal, and gastric cancers. The potency of our cMet ADC is independent of MET gene copy number, and its antitumor activity was high not only in high cMet-expressing cell lines but also in medium-to-low cMet cell lines (40 000-90 000 cMet/cell) in which a cMet ADC with tubulin inhibitor payload was considerably less potent. In vivo xenografts with low-medium cMet expression were also very responsive to TR1801-ADC at a single dose, while a cMet ADC using a tubulin inhibitor showed a substantially reduced efficacy. Furthermore, TR1801-ADC had excellent efficacy with significant antitumor activity in 90% of tested patient-derived xenograft models of gastric, colorectal, and head and neck cancers: 7 of 10 gastric models, 4 of 10 colorectal cancer models, and 3 of 10 head and neck cancer models showed complete tumor regression after a single-dose administration. Altogether, TR1801-ADC is a new generation cMet ADC with best-in-class preclinical efficacy and good tolerability in rats.


Subject(s)
Antineoplastic Agents/pharmacology , Benzodiazepines/pharmacology , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Oncogenes/immunology , Proto-Oncogene Proteins c-met/immunology , Pyrroles/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Biliary Tract Neoplasms/metabolism , Cell Line, Tumor , Colonic Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/metabolism , Humans , Immunoconjugates/therapeutic use , Immunoconjugates/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/immunology , Proto-Oncogene Proteins c-met/metabolism , Rats , Rats, Sprague-Dawley , Stomach Neoplasms/metabolism , Tissue Array Analysis , Xenograft Model Antitumor Assays
10.
Exp Cell Res ; 384(2): 111634, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31541617

ABSTRACT

Great attention has been attached to explore the association between oral bacteria and oral cancer. Recently, four common inhabitants of oral cavity, Porphyromonas gingivalis, Fusobacterium nucleatum, Treponema denticola and Streptococcus anginosus, have been identified as potential etiologic bacterial agents for oral carcinogenesis. They might promote the oncogenesis and progression of oral cancer by induction of chronic inflammation, enhancement of migration and invasiveness, inhibition of cell apoptosis, augment of cell proliferation, suppression of immune system and production of carcinogenic substances. Thus, this review will focus on the possible mechanisms of these oral bacteria contributing to occurrence and development of oral cancer, and the potential clinical implications of utilizing oral bacteria on the diagnosis, prevention and treatment of oral cancer will be discussed.


Subject(s)
Mouth Neoplasms/immunology , Mouth Neoplasms/microbiology , Animals , Bacteria/immunology , Carcinogenesis/immunology , Cell Proliferation/physiology , Humans , Oncogenes/immunology
11.
Nat Rev Immunol ; 19(5): 337-348, 2019 05.
Article in English | MEDLINE | ID: mdl-30890785

ABSTRACT

B cells face multiple restrictions on glucose and energy metabolism. Their lineage-determining transcription factors repress glucose uptake and pentose phosphate pathway activity, while their low numbers of mitochondria and small cytoplasmic volume set narrow limits for mitochondrial ATP production and autophagy as alternative energy sources. During activation, B cells can balance temporary increases of energy expenditure. However, permanent hyperactivation of kinases, for instance, downstream of an autoreactive B cell receptor (BCR) or a transforming oncogene, can cause energy stress and cell death. Here, I propose that B cell-intrinsic restriction of ATP represents a safeguard to eliminate autoreactive or pre-malignant B cells. If the metabolic gatekeepers are compromised, influx of additional glucose may fuel permanent increases in metabolic demands and pathological B cell proliferation, driven by an autoreactive BCR or a transforming oncogene.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , Cell Transformation, Neoplastic/immunology , Oncogenes/immunology , Animals , Cell Proliferation/physiology , Energy Metabolism/immunology , Humans , Mitochondria/immunology
12.
Med Sci Monit ; 25: 888-892, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30739906

ABSTRACT

MYH9 was first discovered due to thrombocytopenia caused by MYH9 mutation-related abnormalities. In recent years, researchers have increasingly found that MYH9 plays an important role in cancer as a cytokine involved in cytoskeletal reorganization, cellular pseudopodia formation, and migration. MYH9 is closely related to the progress and poor prognosis of most solid tumors, and it is now accepted that MYH9 is a suppressor gene and plays an important role on the re-Rho pathway. Recent research has been limited to the study of tissues. However, it would be more direct and informative to be able to use hematology to assess tumor prognosis and changes in MYH9 levels and NMMHC-IIA. This article summarizes recent research on MYH9 and provides a reference for future clinical research.


Subject(s)
Molecular Motor Proteins/metabolism , Molecular Motor Proteins/physiology , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/physiology , Blood Platelets , Genes, Tumor Suppressor/physiology , Humans , Molecular Motor Proteins/genetics , Mutation , Myosin Heavy Chains/genetics , Oncogenes/immunology , Oncogenes/physiology , Thrombocytopenia
13.
JCI Insight ; 3(19)2018 10 04.
Article in English | MEDLINE | ID: mdl-30282837

ABSTRACT

Adoptive cell transfer (ACT) of tumor-infiltrating lymphocytes (TILs) targeting neoantigens can mediate tumor regression in selected patients with metastatic epithelial cancer. However, effectively identifying and harnessing neoantigen-reactive T cells for patient treatment remains a challenge and it is unknown whether current methods to detect neoantigen-reactive T cells are missing potentially clinically relevant neoantigen reactivities. We thus investigated whether the detection of neoantigen-reactive TILs could be enhanced by enriching T cells that express PD-1 and/or T cell activation markers followed by microwell culturing to avoid overgrowth of nonreactive T cells. In 6 patients with metastatic epithelial cancer, this method led to the detection of CD4+ and CD8+ T cells targeting 18 and 1 neoantigens, respectively, compared with 6 and 2 neoantigens recognized by CD4+ and CD8+ T cells, respectively, when using our standard TIL fragment screening approach. In 2 patients, no recognition of mutated peptides was observed using our conventional screen, while our high-throughput approach led to the identification of 5 neoantigen-reactive T cell receptors (TCRs) against 5 different mutations from one patient and a highly potent MHC class II-restricted KRASG12V-reactive TCR from a second patient. In addition, in a metastatic tumor sample from a patient with serous ovarian cancer, we isolated 3 MHC class II-restricted TCRs targeting the TP53G245S hot-spot mutation. In conclusion, this approach provides a highly sensitive platform to isolate clinically relevant neoantigen-reactive T cells or their TCRs for cancer treatment.


Subject(s)
Cell Separation/methods , Flow Cytometry/methods , Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/transplantation , Neoplasms/therapy , Adult , Aged , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis/genetics , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Mutation , Neoplasms/genetics , Neoplasms/immunology , Oncogenes/genetics , Oncogenes/immunology , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Sensitivity and Specificity , Tumor Cells, Cultured
14.
Nat Rev Cancer ; 18(3): 139-147, 2018 03.
Article in English | MEDLINE | ID: mdl-29326431

ABSTRACT

Immunotherapeutic interventions are showing effectiveness across a wide range of cancer types, but only a subset of patients shows clinical response to therapy. Responsiveness to checkpoint blockade immunotherapy is favoured by the presence of a local, CD8+ T cell-based immune response within the tumour microenvironment. As molecular analyses of tumours containing or lacking a productive CD8+ T cell infiltrate are being pursued, increasing evidence is indicating that activation of oncogenic pathways in tumour cells can impair induction or execution of a local antitumour immune response. This Review summarizes our current knowledge of the influence of oncogenic effects on evasion of antitumour immunity.


Subject(s)
Neoplasms/immunology , Neoplasms/therapy , Oncogenes/immunology , Tumor Escape , AMP-Activated Protein Kinase Kinases , CD8-Positive T-Lymphocytes/drug effects , Genes, myc/immunology , Humans , Immunotherapy/methods , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Oncogenes/physiology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , PTEN Phosphohydrolase/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway
15.
Enferm. infecc. microbiol. clín. (Ed. impr.) ; 35(9): 593-602, nov. 2017. ilus, tab, graf
Article in Spanish | IBECS | ID: ibc-168888

ABSTRACT

La infección por el virus del papiloma humano (VPH) es la infección de transmisión sexual más frecuente en el mundo. Este virus ocasiona generalmente lesiones benignas, como verrugas genitales, pero también su persistencia ocasiona procesos malignos, como cáncer de cuello de útero (CCU) y, menos frecuentemente, anal, vaginal y de la cavidad orofaríngea. El CCU es una enfermedad muy severa, con alta mortalidad en muchos países. El cribado de CCU con citología ha tenido mucho éxito en estos últimos años, pero hay innumerable evidencia científica para que sea sustituida por la detección del VPH como prueba inicial. Para esto, hay en el mercado gran cantidad de técnicas, siendo aconsejable utilizar sistemas automáticos y pruebas aprobadas por la FDA. Un nuevo algoritmo basado en la detección individualizada de los genotipos 16 y 18 presentes en el 70% de los CCU ha sido propuesto por expertos y su implantación será inmediata en algunos países (AU)


Infection with human papillomavirus (HPV) is the leading cause of sexually transmitted infection worldwide. This virus generally causes benign lesions, such as genital warts, but persistent infection may lead to cervical cancer, anal cancer, vaginal cancer, and oropharyngeal cancer, although less frequently. Cervical cancer is a severe disease with a high mortality in some countries. Screening with cytology has been very successful in the last few years, but nowadays there are numerous studies that confirm that cytology should be replaced with the detection of HPV as a first line test in population based screening. There are several commercially available FDA approved tests for screening of cervical cancer. A new strategy, based on individual detection of the high risk genotypes HPV16 and HPV18, present in 70% of cervical cancer biopsies, has been proposed by some experts, and is going to be implemented in most countries in the future (AU)


Subject(s)
Humans , Papillomavirus Infections/microbiology , Papillomaviridae/isolation & purification , Uterine Cervical Neoplasms/microbiology , Microbiological Techniques/methods , Mass Screening/methods , Oncogenes/immunology , Biomarkers, Tumor/analysis
16.
Expert Opin Biol Ther ; 17(5): 595-611, 2017 05.
Article in English | MEDLINE | ID: mdl-28330383

ABSTRACT

INTRODUCTION: The possibility of correcting defective genes and modulating gene expression through gene therapy has emerged as a promising treatment strategy for breast cancer. Furthermore, the relevance of tumor immune microenvironment in supporting the oncogenic process has paved the way for novel immunomodulatory applications of gene therapy. Areas covered: In this review, the authors describe the most relevant delivery systems, focusing on nonviral vectors, along with the description of the major approaches used to modify target cells, including gene transfer, RNA interference (RNAi), and epigenetic regulation. Furthermore, they highlight innovative therapeutic strategies and the application of gene therapy in clinical trials for breast cancer. Expert opinion: Gene therapy has the potential to impact breast cancer research. Further efforts are required to increase the clinical application of RNAi-based therapeutics, especially in combination with conventional treatments. Innovative strategies, including genome editing and stem cell-based systems, may contribute to translate gene therapy into clinical practice. Immune-based approaches have emerged as an attractive therapeutic opportunity for selected breast cancer patients. However, several challenges need to be addressed before considering gene therapy as an actual option for the treatment of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/therapy , Genetic Therapy/trends , Animals , Breast Neoplasms/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Clinical Trials as Topic/methods , Epigenesis, Genetic/genetics , Epigenesis, Genetic/immunology , Female , Genetic Therapy/methods , Humans , Oncogenes/genetics , Oncogenes/immunology , RNA Interference/physiology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
17.
Nat Rev Clin Oncol ; 14(2): 115-128, 2017 02.
Article in English | MEDLINE | ID: mdl-27245281

ABSTRACT

Telomerase reverse transcriptase (TERT) is a self-antigen that is expressed constitutively in many tumours, and is, therefore, an important target for anticancer immunotherapy. In the past 10 years, trials of immunotherapy with TERT-based vaccines have demonstrated only modest benefits. In this Perspectives, I discuss the possible immunological reasons for this limited antitumour efficacy, and propose that advances in our understanding of the genetics and biology of the involvement of TERT in cancer provides the basis for renewed interest in TERT- based immunotherapy. Telomerase and TERT are expressed in cancer cells at every stage of tumour evolution, from the cancer stem cell to circulating tumour cells and tumour metastases. Many cancer types also harbour cells with mutations in the TERT promoter region, which increase transcriptional activation of this gene. These new findings should spur new interest in the development of TERT-based immunotherapies that are redesigned in line with established immunological considerations and working principles, and are tailored to patients stratified on the basis of TERT-promoter mutations and other underlying tumour characteristics. Thus, despite the disappointment of previous clinical trials, TERT offers the potential for personalized immunotherapy, perhaps in combination with immune-checkpoint inhibition.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Telomerase/therapeutic use , Autoimmunity/immunology , Cancer Vaccines/immunology , Clinical Trials as Topic , Epithelial-Mesenchymal Transition/immunology , Humans , Immunity, Cellular/drug effects , Neoplasms/enzymology , Neoplasms/immunology , Neoplastic Cells, Circulating/immunology , Oncogenes/immunology , Promoter Regions, Genetic/genetics , Telomerase/immunology
18.
Cytokine ; 89: 209-218, 2017 01.
Article in English | MEDLINE | ID: mdl-26631911

ABSTRACT

Tyrosine kinase 2 (TYK2) is a member of the Janus kinase (JAK) family, which transduces cytokine and growth factor signalling. Analysis of TYK2 loss-of-function revealed its important role in immunity to infection, (auto-) immunity and (auto-) inflammation. TYK2-deficient patients unravelled high similarity between mice and men with respect to cellular signalling functions and basic immunology. Genome-wide association studies link TYK2 to several autoimmune and inflammatory diseases as well as carcinogenesis. Due to its cytokine signalling functions TYK2 was found to be essential in tumour surveillance. Lately TYK2 activating mutants and fusion proteins were detected in patients diagnosed with leukaemic diseases suggesting that TYK2 is a potent oncogene. Here we review the cell intrinsic and extrinsic functions of TYK2 in the characteristics preventing and enabling carcinogenesis. In addition we describe an unexpected function of kinase-inactive TYK2 in tumour rejection.


Subject(s)
Leukemia/immunology , Mutation , Neoplasm Proteins/immunology , Oncogenes/immunology , Signal Transduction/immunology , TYK2 Kinase/immunology , Animals , Genome-Wide Association Study , Humans , Leukemia/genetics , Mice , Neoplasm Proteins/genetics , Signal Transduction/genetics , TYK2 Kinase/genetics
19.
Int J Radiat Biol ; 92(10): 577-82, 2016 10.
Article in English | MEDLINE | ID: mdl-27548028

ABSTRACT

PURPOSE: Cancer initialization can be explained as a result of parasitic virus energy consumption leading to randomized genome chemical bonding. MATERIALS AND METHODS: Analysis of experimental data on cell-mediated immunity (CMI) containing about 12,000 cases of healthy humans, cancer patients and patients with precancerous cervical lesions disclosed that the specific cancer and the non-specific lactate dehydrogenase-elevating (LDH) virus antigen elicit similar responses. The specific antigen is effective only in cancer type of its origin but the non-specific antigen in all examined cancers. CMI results of CIN patients display both healthy and cancer state. The ribonucleic acid (RNA) of the LDH virus parasitizing on energy reduces the ratio of coherent/random oscillations. Decreased effect of coherent cellular electromagnetic field on bonding electrons in biological macromolecules leads to elevating probability of random genome reactions. RESULTS: Overlapping of wave functions in biological macromolecules depends on energy of the cellular electromagnetic field which supplies energy to bonding electrons for selective chemical bonds. CMI responses of cancer and LDH virus antigens in all examined healthy, precancerous and cancer cases point to energy mechanism in cancer initiation. CONCLUSIONS: Dependence of the rate of biochemical reactions on biological electromagnetic field explains yet unknown mechanism of genome mutation.


Subject(s)
Electromagnetic Fields , Mutation/genetics , Neoplasms/genetics , Neoplasms/immunology , Oncogenes/genetics , Oncogenes/immunology , Computer Simulation , Humans , Lactate dehydrogenase-elevating virus/physiology , Models, Chemical , Models, Genetic , Models, Immunological , Mutation/radiation effects , Neoplasms/virology , Oncogenes/radiation effects , T-Lymphocytes/immunology , T-Lymphocytes/radiation effects , T-Lymphocytes/virology
20.
Immunol Rev ; 253(1): 158-66, 2013 May.
Article in English | MEDLINE | ID: mdl-23550645

ABSTRACT

Neoplastic transformation is caused by accumulation of genetic lesions that ultimately convert normal cells into tumor cells with uncontrolled proliferation and survival, unlimited replicative potential, and invasive growth. Emerging evidence has highlighted the functional importance of non-coding RNAs, particularly microRNAs (miRNAs), in the initiation and progression of tumor development. The mir-17-92 miRNA is among the best characterized miRNA oncogenes, whose genomic amplification or aberrant elevation are frequently observed in a variety of tumor types. Unlike protein-coding oncogenes, where one transcript produces one protein, mir-17-92 encodes a polycistronic miRNA transcript that yields six individual miRNA components. This unique gene structure, shared by many important miRNA oncogenes and tumor suppressors, underlies the unique functionality of mir-17-92 in a cell type and context-dependent manner. Recent functional dissection of mir-17-92 indicates that individual mir-17-92 components perform distinct biological functions, which collectively regulate multiple related cellular processes during development and disease. The structural complexity of mir-17-92 as a polycistronic miRNA oncogene, along with the complex mode of interactions among its components, constitutes the molecular basis for its unique functional complexity during normal and tumor development.


Subject(s)
Cell Transformation, Neoplastic , MicroRNAs/immunology , Neoplasms/immunology , Oncogenes/immunology , Animals , Cell Transformation, Neoplastic/genetics , Cellular Microenvironment , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/biosynthesis , Neoplasms/genetics , Organ Specificity , RNA, Long Noncoding , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...