Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Ther Adv Med Oncol ; 16: 17588359231220516, 2024.
Article in English | MEDLINE | ID: mdl-38188467

ABSTRACT

Background: Immune checkpoint inhibitor therapy has demonstrated impressive clinical benefits in multiple tumor types. TQB2450, a novel monoclonal antibody targeting programmed cell death ligand 1, has shown safety and efficacy in preclinical studies. Objectives: This first-in-human study aimed to evaluate the safety/tolerability, pharmacokinetics (PK), immunogenicity, and preliminary antitumor activity of TQB2450 in patients with advanced malignant tumors. Design and methods: In this phase I study, eligible patients with advanced malignant tumors received intravenous TQB2450 once every 3 weeks. This study consisted of a 3 + 3 dose-escalation phase (1-30 mg/kg) and a specific dose-expansion phase (1200 mg). The primary endpoints were maximum tolerated dose (MTD), dose-limiting toxicity (DLT), and safety. The secondary endpoints were PK, immunogenicity, and investigator-assessed response rate. Results: Between April 2018 and February 2020, 40 patients were enrolled (22 in the dose-escalation phase and 18 in the dose-expansion phase). No DLT was reported and the MTD was not reached. Grade ⩾3 or worse treatment-related treatment-emergent adverse events (AEs) occurred in 11 (27.50%) patients, with the most frequent being aspartate aminotransferase increased (5.00%), leukopenia (5.00%), and anemia (5.00%). Treatment-related serious AEs were reported in six patients, the most common of which was decompensated liver function (5.00%). No treatment-related death was reported. The maximum serum concentration of TQB2450 increased in a dose-proportional manner. Treatment-induced anti-drug antibodies were detected in 31.58% (12/38) of patients. The investigator assessed the objective response rate as 5.00% and the disease control rate was 52.50%, including 2 partial responses and 19 stable diseases. The median progression-free survival was 2.69 (95% confidence interval, 2.07-6.14) months. Conclusion: TQB2450 has a manageable safety profile with favorable PK and immunogenicity and has shown early evidence of clinical activity in advanced malignant tumors. ClinicalTrialsgov identifier: NCT03460457.

2.
Front Oncol ; 13: 1276009, 2023.
Article in English | MEDLINE | ID: mdl-37936609

ABSTRACT

Background: This study aimed to identify potential biomarkers in patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC) and further probe the prognostic implications of CDKN2A mutations, particularly within a subset receiving immunotherapy. Methods: In this retrospective single-center study, we evaluated the next-generation sequencing (NGS) data from Foundation Medicine (FM) for patients with recurrent or metastatic HNSCC between January 1, 2019, and December 31, 2021. Patients were stratified based on CDKN2A loss-of-function (LOF) versus wild-type (WT) categorizations, with a focused subgroup analysis on those administered immunotherapy. Results: The study encompassed 77 patients, of which 62 had undergone immunotherapy. The median duration of follow-up was 22.6 months. For the CDKN2A LOF group, the median overall survival (OS) was 16.5 months, contrasted with 30.0 months in the CDKN2A WT group (P=0.014). Notably, female gender (hazard ratio [HR]=4.526, 95% confidence interval [CI]: 1.934-10.180, P=0.0003) and CDKN2A LOF (HR=2.311, 95% CI: 1.156-4.748, P=0.019) emerged as independent risk factors for mortality in patients with recurrent or metastatic HNSCC. Within the immunotherapy subset, the median OS was 11.7 months for the CDKN2A LOF group, and 22.5 months for the CDKN2A WT group (P=0.017). Further, the female gender (HR=4.022, 95% CI: 1.417-10.710, P=0.006), CDKN2A LOF (HR=4.389, 95% CI: 1.782-11.460, P=0.002), and a combined positive score below 1 (HR=17.20, 95% CI: 4.134-79.550, P<0.0001) were identified as significant predictors of mortality among patients with recurrent or metastatic HNSCC receiving immunotherapy. Conclusion: Alterations manifesting as LOF in the CDKN2A gene stand as robust indicators of unfavorable survival outcomes in HNSCC patients, including the subset that underwent immunotherapy.

3.
Front Oncol ; 12: 971594, 2022.
Article in English | MEDLINE | ID: mdl-35992822

ABSTRACT

Bone metastases are common complications of solid tumors. The outcome is poor despite major progress in cancer therapies. We describe a multicenter, open-label, phase 1, dose escalation and expansion trial of JMT103, a novel fully humanized receptor activator of nuclear factor kappa-B ligand (RANKL)-targeting monoclonal antibody, in adults with bone metastases from solid tumors. The study assessed the safety, tolerability, and pharmacokinetics/pharmacodynamics of JMT103. Patients received JMT103 at doses of 0.5, 1.0, 2.0, and 3.0 mg/kg every 4 weeks for 3 cycles. Among 59 patients enrolled, 20 and 39 patients participated in the dose-escalation and dose-expansion phases, respectively. One dose-limiting toxicity was observed at 2.0 mg/kg. The maximum tolerated dose was not determined. Treatment-related adverse events were reported in 29 (49.2%) patients, most commonly hypophosphatemia (30.5%), hypocalcemia (23.7%), and hypermagnesemia (10.2%). No treatment-related serious adverse events were reported. Two patients died due to disease progression, which were attributed to gastric cancer and lung neoplasm malignant respectively. Dose proportionality occurred between exposure levels and administered dose was within a dose range of 0.5 to 3.0 mg/kg. The suppression of urinary N-telopeptide corrected for creatinine was rapid, significant, and sustained across all doses of JMT103, with the median change from baseline ranging from -61.4% to -92.2% at day 141. JMT103 was well tolerated in patients with bone metastases from solid tumors, with a manageable safety profile. Bone antiresorptive activity shows the potential of JMT103 for treatment of bone metastases from solid tumors. Registration No.: NCT03550508; URL: https://www.clinicaltrials.gov/.

4.
Head Neck ; 44(9): 2040-2045, 2022 09.
Article in English | MEDLINE | ID: mdl-35915862

ABSTRACT

BACKGROUND: There is limited evidence supporting the use of taxane-based chemotherapy combined with cetuximab to treat recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC). This retrospective study aimed to explore treatment efficacy and safety in a first-line setting. METHODS: Fifty-nine patients received ≤6 cycles of combined cetuximab, platinum compounds, and taxane (median follow-up, 352 days). Kaplan-Meier survival curves were constructed. RESULTS: The median patient age was 54 years (23-75 years; 50 males and 9 females). The most common distant metastatic site was the lung. Patients received ≥2 cycles chemotherapy (33 [55.9%] received cetuximab, paclitaxel, and carboplatin; 21 [35.5%] received cetuximab maintenance; median progression-free survival, 7 months; overall survival, 12 months). The most common hematological toxicity was Grade 3 or 4 neutropenia, which was successfully managed through growth-stimulating factors and dose modifications. No treatment-associated deaths occurred. CONCLUSIONS: Combined cetuximab, platinum, and taxane is effective and tolerable in R/M HNSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/pathology , Cetuximab/therapeutic use , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/etiology , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/etiology , Platinum/therapeutic use , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/etiology , Taxoids/therapeutic use
5.
JAMA Oncol ; 8(7): 1042-1046, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35511148

ABSTRACT

Importance: The antibody drug conjugate drug MRG003 comprises an anti-epidermal growth factor receptor (EGFR) humanized immunoglobulin G1 monoclonal antibody that is conjugated with monomethyl auristatin E via a valine-citrulline linker. There is currently insufficient evidence of this drug's safety and efficacy. Objective: To evaluate the safety and maximum tolerated dose of MRG003 in a phase 1a study and investigate the preliminary antitumor activity in EGFR-expressing patients in a phase 1b study. Design, Setting, and Participants: This nonrandomized open-label, single-arm, phase 1, multicenter study of solid tumors was divided into 2 parts, phase 1a dose escalation and phase 1b dose expansion. Patients with advanced or metastatic solid tumors who had failed outcomes from or were not able to receive standard treatment were enrolled in phase 1a without EGFR prescreening. Phase 1b recruited EGFR-positive patients with refractory advanced squamous cell carcinomas of the head and neck (SCCHN), nasopharyngeal carcinoma (NPC), and colorectal cancer (CRC). This study was conducted at 7 Chinese centers between April 11, 2018, and March 29, 2021 (data cutoff date). Data analysis took place between April 2021 and June 2021. Interventions: An intravenous dose of 0.1 to 2.5 mg/kg of MRG003 was administered every 3 weeks during phase 1a. During phase 1b, patients were administered the recommended dose identified in phase 1a. Main Outcomes and Measures: The primary end points were dose-limiting toxic effects in phase 1a and objective response rate in phase 1b. The safety, tolerability, immunogenicity, and pharmacokinetics of MRG003 were assessed. Tumor assessment was evaluated by RECIST 1.1. Results: Twenty-two patients (mean [range] age, 54.5 [32.0-67.0] years; 9 women [41%]) were enrolled in phase 1a and 39 patients (mean [range] age, 50.4 [27.0-75.0] years; 8 women [21%]) in phase 1b. The recommended dose was identified as 2.5 mg/kg. Eighty-nine percent of adverse events (AEs) were associated with MRG003 treatment, and most AEs were grade 1 to 2. Nineteen patients (31%) reported grade 3 or greater treatment-related AEs, including hyponatremia, leukocytopenia, neutropenia, increased aspartate aminotransferase levels, and febrile neutropenia. In phase 1a, 1 patient (5%) achieved a partial response, and 5 (23%) achieved stable disease. In phase 1b, 8 patients (21%) achieved a confirmed partial response, and 12 (31%) achieved stable disease. The objective response rates for SCCHN, NPC, and CRC were 40%, 44%, and 0%, and the disease control rates were 100%, 89%, and 25%, respectively. Conclusions and Relevance: The findings of this nonrandomized clinical trial suggest that MRG003 showed a manageable safety profile and promising antitumor activity in patients with EGFR-positive NPC and SCCHN. Trial Registration: Clinicaltrials.gov Identifier: NCT04868344.


Subject(s)
Antibodies, Monoclonal, Humanized , Immunoconjugates , Neoplasms , Antibodies, Monoclonal, Humanized/adverse effects , ErbB Receptors , Female , Humans , Immunoconjugates/adverse effects , Middle Aged , Neoplasms/drug therapy , Receptors, Growth Factor
6.
J Immunol Res ; 2020: 1786505, 2020.
Article in English | MEDLINE | ID: mdl-33062718

ABSTRACT

[This corrects the article DOI: 10.1155/2019/3690561.].

8.
Aging (Albany NY) ; 12(13): 13701-13715, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32629428

ABSTRACT

Lung cancer is the leading cause of cancer death worldwide and accounts for more than one-third of all newly diagnosed cancer cases in China. Therefore, it is of great clinical significance to explore new driver gene mutations in non-small-cell lung cancer (NSCLC). Using an initial bioinformatic analysis, we identified somatic gene mutations in 13 patients with NSCLC and confirmed these mutations by targeted sequencing in an extended validation group of 88 patients. Recurrent mutations were detected in UNC5D (7.9%), PREX1 (5.0%), HECW1 (4.0%), DACH1 (2.0%), and GPC5 (2.0%). A functional study was also performed in UNC5D mutants. Mutations in UNC5D promoted tumorigenesis by abolishing the tumor suppressor function of the encoded protein. Additionally, in ten patients with lung squamous cell carcinoma, we identified mutations in KEAP1/NFE2L2 that influenced the expression of target genes in vivo and in vitro. Overall, the results of our study expanded the known spectrum of driver mutations involved in the pathogenesis of NSCLC.


Subject(s)
Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Adult , Aged , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Case-Control Studies , Computational Biology , DNA Mutational Analysis , Humans , Lung/pathology , Lung/surgery , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Mice , Middle Aged , Mutation , Pneumonectomy , Exome Sequencing , Xenograft Model Antitumor Assays
9.
J Immunol Res ; 2019: 3690561, 2019.
Article in English | MEDLINE | ID: mdl-31781676

ABSTRACT

Colorectal cancer (CRC) remains the third most commonly diagnosed cancer, ranking second among the most common causes of cancer-related mortality. Immune checkpoint therapy has recently been shown to have great potential. However, only some patients respond to immune checkpoint blockade, indicating the unmet need for determining the underlying mechanism of colorectal cancer immunosuppression. In this study, we analyzed The Cancer Genome Atlas (TCGA) datasets and found that high expression of PIPKIγ positively correlated with tumor-associated macrophage infiltration. Further loss-of-function studies revealed that silencing PIPKIγ greatly reduced CCL2 expression at both the mRNA and protein levels, leading to weak chemotaxis of cancer cells to macrophages. Mechanistically, PIPKIγ facilitated PI3K-Akt-mTOR signaling pathway activation to increase STAT3 phosphorylation levels, thus triggering CCL2 transcription to enhance tumor-associated macrophage recruitment. These findings identify the PIPKIγ signaling pathway as a new actor in colorectal cancer immunosuppression and a potential therapeutic target for this common cancer.


Subject(s)
Chemokine CCL2/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
10.
BMC Cancer ; 19(1): 1094, 2019 Nov 12.
Article in English | MEDLINE | ID: mdl-31718595

ABSTRACT

BACKGROUND: The incidence of papillary thyroid carcinoma (PTC) has been increasing worldwide in recent years. Therefore, novel potential therapeutic targets for PTC are urgently needed. Enhancer of zeste homolog 2 (EZH2), a methyltransferase belonging to PRC2, plays important roles in epigenetic silencing and cell cycle regulation. EZH2 overexpression has been found in several malignant tumor tissues, while its expression and function in PTC are largely unknown. METHODS: Sixty-five cases of PTC tissue confirmed by pathology and 30 cases of normal thyroid tissue adjacent to PTC tissue were collected from patients undergoing surgical treatment, between February 2003 and February 2006. We investigated the clinic pathologic significance of EZH2 expression using Realtime-PCR and IHC in 65 human PTC tissues and 30 normal thyroid tissue samples. The EZH2 expression in human PTC cell lines (K1 and W3) and the normal thyroid follicular epithelial cell line Nthy-ori 3-1 was analyzed by Western blotting and Realtime PCR. The expressions of ERα and ERß in cell lines were analyzed by Realtime PCR.The tumor cell biological behavior was evaluated by CCK8 assay, colony formation assay, transwell migration assay and xenograft tumors model. RESULTS: Higher rate of EZH2 expression was found in PTC tissues than in normal thyroid tissues, EZH2 expression is associated with lymph node metastasis and recurrent. Inhibition of EZH2 in PTC cell lines downregulates cellular proliferation and migration. PTC is a disease with high incidence of female and E2-ERα upregulates EZH2 expression. CONCLUSIONS: These results suggest a potential role of EZH2 for the PTC growth and metastasis. As a novel therapy, a pharmacological therapy targeting EZH2 has full potential in treatment of PTC.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/metabolism , Adult , Aged , Animals , Case-Control Studies , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Heterografts , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Thyroid Cancer, Papillary/pathology , Tumor Burden
11.
JAMA Netw Open ; 2(5): e193348, 2019 05 03.
Article in English | MEDLINE | ID: mdl-31050781

ABSTRACT

Importance: Thyrotoxic periodic paralysis (TPP) is a potentially lethal complication of hyperthyroidism. However, only 1 specific susceptibility locus for TPP has been identified. Additional genetic determinants should be detected so that a prediction model can be constructed. Objective: To investigate the genetic architecture of TPP and distinguish TPP from Graves disease cohorts. Design, Setting, and Participants: This population-based case-control study used a 2-stage genome-wide association study to investigate the risk loci of TPP and weighted genetic risk score to construct a TPP prediction model with data from a Chinese Han population recruited in hospitals in China from March 2003 to December 2015. The analysis was conducted from November 2014 to August 2016. Main Outcomes and Measures: Loci specifically associated with TPP risk and those shared with Graves disease and prediction model of joint effects of TPP-specific loci. Results: A total of 537 patients with TPP (mean [SD] age, 35 [11] years; 458 male) 1519 patients with Graves disease and no history of TPP (mean [SD] age, 38 [13] years; 366 male), and 3249 healthy participants (mean [SD] age, 46 [10] years; 1648 male) were recruited from the Han population by hospitals throughout China. Two new TPP-specific susceptibility loci were identified: DCHS2 on 4q31.3 (rs1352714: odds ratio [OR], 1.58; 95% CI, 1.35-1.85; P = 1.24 × 10-8) and C11orf67 on 11q14.1 (rs2186564: OR, 1.50; 95% CI, 1.29-1.74; P = 2.80 × 10-7). One previously reported specific locus was confirmed on 17q24.3 near KCNJ2 (rs312729: OR, 2.08; 95% CI, 1.83-2.38; P = 8.02 × 10-29). Meanwhile, 2 risk loci (MHC and Xq21.1) were shared by Graves disease and TPP. After 2 years of treatment, the ratio of persistent thyrotropin receptor antibody positivity was higher in patients with TPP than in patients with Graves disease and no history of TPP (OR, 3.82; 95% CI, 2.04-7.16; P = 7.05 × 10-6). The prediction model using a weighted genetic risk score and 11 candidate TPP-specific single-nucleotide polymorphisms had an area under the curve of 0.80. Conclusions and Relevance: These findings provide evidence that TPP is a novel molecular subtype of Graves disease. The newly identified loci, along with other previously reported loci, demonstrate the growing complexity of the heritable contribution to TPP pathogenesis. A complete genetic architecture will be helpful to understand the pathophysiology of TPP, and a useful prediction model could prevent the onset of TPP.


Subject(s)
Graves Disease/genetics , Thyroid Crisis/genetics , Adult , Asian People/genetics , Case-Control Studies , China , Cross-Sectional Studies , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Paralysis/genetics , Polymorphism, Single Nucleotide
12.
Biomed Pharmacother ; 115: 108921, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31078045

ABSTRACT

Colorectal cancer is the third most common cancer worldwide and shows resistance to immune checkpoint inhibitors which have been demonstrated to be effective in many other types of cancers. Pre-existing T-cell response in tumor microenvironment often determines the therapeutic benefit of immune checkpoint blockade. Tumor-infiltrating CD8+ T-cells are considered as the major effector immune cells in antitumor immunity. In this study, we aimed to identify the intrinsic oncogenic pathway that contributes to a reduction of CD8+ T-cell infiltration in colorectal cancer. To achieve this, human colon adenocarcinoma samples derived from The Cancer Genome Altas (TCGA) were stratified into low T-cell-inflamed and high T-cell-inflamed groups based on the expression of T-cell signature genes. Gene set enrichment analysis of revealed a close correlation between activation of the Wnt/ß-catenin signaling pathway and absence of T-cell infiltration. By immunohistochemical analysis of 155 colorectal cancer tissues, we found that tumors with high ß-catenin expression showed a significant reduction of CD8+ T-cell infiltration. Mechanistically, ß-catenin can regulate CCL4 expression to recruit CD103+ dendritic cells to enable CD8+ T cell activation. Collectively, our data indicate that oncogenic ß-catenin signal may mediate colorectal cancer resistance to immunotherapies, pointing to the combined PD-1-immunotherapy with targeting ß-catenin in colorectal cancer.


Subject(s)
Adenocarcinoma/immunology , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Wnt Signaling Pathway/immunology , beta Catenin/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Chemokine CCL4/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Immunotherapy , Lymphocyte Activation/genetics , Signal Transduction/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Wnt Signaling Pathway/genetics , beta Catenin/genetics
13.
EBioMedicine ; 44: 375-386, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31105034

ABSTRACT

BACKGROUND: Emerging evidence suggests that metabolic alterations are a hallmark of cancer cells and contribute to tumor initiation and development. Cancer cells primarily utilize aerobic glycolysis (the Warburg effect) to produce energy and support anabolic growth. The type Iγ phosphatidylinositol phosphate kinase (PIPKIγ) is profoundly implicated in tumorigenesis, however, little is known about its role in reprogrammed energy metabolism. METHODS: Loss- and gain-of-function studies were applied to determine the oncogenic roles of PIPKIγ in colorectal cancer. Transcriptome analysis, real-time qPCR, immunohistochemical staining, Western blotting, and metabolic analysis were carried out to uncover the cellular mechanism of PIPKIγ. FINDINGS: In this study, we showed that PIPKIγ was frequently upregulated in colorectal cancer and predicted a poor prognosis. Genetic silencing of pan-PIPKIγ suppressed cell proliferation and aerobic glycolysis of colorectal cancer. In contrast, the opposite effects were observed by overexpression of PIPKIγ_i2. Importantly, PIPKIγ-induced prolific effect was largely glycolysis-dependent. Mechanistically, PIPKIγ facilitated activation of PI3K/Akt/mTOR signaling pathways to upregulate c-Myc and HIF1α levels, which regulate expression of glycolytic enzymes to enhance glycolysis. Moreover, pharmacological inhibition by PIPKIγ activity with the specific inhibitor UNC3230 significantly inhibited colorectal cancer glycolysis and tumor growth. INTERPRETATION: Our findings reveal a new regulatory role of PIPKIγ in Warburg effect and provide a key contributor in colorectal cancer metabolism with potential therapeutic potentials. FUND: National Key Research and Development Program of China, Outstanding Clinical Discipline Project of Shanghai Pudong, Natural Science Foundation of China, and Science and Technology Commission of Shanghai Municipality.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Glucose/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Gene Expression , Gene Expression Profiling , Gene Knockout Techniques , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , TOR Serine-Threonine Kinases/metabolism
15.
J Clin Endocrinol Metab ; 102(2): 652-660, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27809695

ABSTRACT

Context: Graves disease (GD) is a common autoimmune disease triggered by genetic predisposition and environmental factors. However, the mechanisms of interaction between genetic and environmental factors contributing to the development of GD remain unknown. Objective: We aimed to identify GD susceptibility variants and genes on Xq21.1 locus and interpret the contribution of interaction between genetic predisposition on Xq21.1 and environmental factors to GD. Design: We performed refining study on Xq21.1 in a 2-stage study and carried out expression quantitative trait locus analysis of the best association signal with GD. Setting and Participants: A total of 4316 GD patients and 4374 sex-matched controls were collected from the Chinese Han population by cooperation with multiple hospitals. Results: We identified that rs3827440 or its linkage single nucleotide polymorphisms (SNPs) were probably the causal variant in the Xq21.1 locus, with the most substantial association with GD in our combined cohorts (P = 2.45 × 10-15). The genotypes of rs3827440 were correlated with the expression of ITM2A in monocytes and peripheral blood mononuclear cells (PBMCs) from healthy volunteers. Notably, the expression of ITM2A in monocytes after lipopolysaccharide (LPS) and interferon-γ (INF-γ) stimulation showed substantial difference among the volunteers that carried different genotypes of rs3827440 (P = 9.40 × 10-7 and P = 1.26 × 10-5 for 24 hours' LPS and INF-γ stimulation, respectively). Moreover, ITM2A expression was significantly decreased in PBMCs from untreated GD patients than that from controls. Conclusion: The results suggest that ITM2A might be a susceptibility gene for GD in the Xq21.1 locus, and environmental factors, such as viral and bacterial infections, probably contribute to GD pathogenesis by interacting with the risk SNP rs3827440 mediating the regulation of ITM2A expression.


Subject(s)
Bacterial Infections/complications , Gene-Environment Interaction , Graves Disease/etiology , Graves Disease/genetics , Leukocytes, Mononuclear/metabolism , Membrane Proteins/genetics , Virus Diseases/complications , China , Genetic Predisposition to Disease , Genome-Wide Association Study , Graves Disease/blood , Humans , Polymorphism, Single Nucleotide , Quantitative Trait Loci
16.
Endocrine ; 53(3): 784-90, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27150612

ABSTRACT

17-hydroxylase deficiency (17OHD) has long been regarded as a rare form of congenital adrenal hyperplasia, inherited in an autosomal recessive pattern. Fifteen patients with 17OHD were described from clinical manifestations and hormone profile. Then, CYP17A1 gene was amplified and sequenced in a new patient. Heterozygous mutations c. 431_433del, p.K144del/c. 985_987delinsAA, p.Y329 fs were identified. Functional study indicated the novel mutation K144del completely abolished enzyme activity. In the three-dimensional model, the K144del mutation completely destroyed the alpha helix in the steroid binding domain. Sixteen SNPs within CYP17A1 gene were selected and genotyped in 7 unrelated families to determine whether Y329 fs had founder effect in China. Haplotyping study showed that all c. 985_987delinsAA mutation shared the same haplotype. However, from GWAS data of 2760 controls, this special haplotype was found only in one chromosome. In conclusion, we identified a novel (K144del) and a widely reported (Y329 fs) heterozygous mutations of CYP17A1 gene from a 17OHD patient. Haplotyping analysis showed the common mutation Y329 fs in China came from the same ancestor, which explains the reason that 17OHD was the second cause of CAH in China.


Subject(s)
Adrenal Hyperplasia, Congenital/genetics , Asian People/genetics , Genotype , Mutation , Steroid 17-alpha-Hydroxylase/genetics , Adolescent , Adult , Child, Preschool , China , DNA Mutational Analysis , Female , Founder Effect , Haplotypes , Humans , Young Adult
17.
Endocrine ; 45(3): 365-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23934357

ABSTRACT

The aim of this study is to assess the association of the SCGB3A2 -112G>A promoter polymorphism with Graves' disease(GD) using a meta-analysis. Relevant studies were identified using PubMed and EMBASE electronic databases. A meta-analysis of relevant studies was performed. This meta-analysis included four case-control studies, containing 6,913 GD cases (Caucasian 3904, Han 3009) and 7,185 controls(Caucasian 4155, Han 3030). The combined results showed a significant difference in genotype distribution (-112A/G) between GD and control populations (A vs. G P = 1.53 × 10(-7); GG vs. AA+AG P = 6.78 × 10(-9)). Meta-analysis was performed using a fixed-effects model. Under the dominant model (GG/AA + GA), the AA and GA genotypes were significantly associated with GD (pooled OR = 1.24, 95 % CI 1.12-1.37). When the two European studies are combined, the AA and GA genotypes were also significantly associated with GD (pooled OR = 1.29, 95 % CI 1.20-1.39). This meta-analysis suggests that SCGB3A2 polymorphism at positions -112G>A was associated with GD both in Chinese and Caucasian population.


Subject(s)
Genetic Predisposition to Disease , Graves Disease/genetics , Polymorphism, Genetic/genetics , Secretoglobins/genetics , Graves Disease/ethnology , Humans
18.
Eur J Endocrinol ; 170(1): 109-19, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24144966

ABSTRACT

BACKGROUND: Convincing evidence has demonstrated the association of TSH receptor (TSHR) with Graves' disease (GD) in the Chinese Han population. OBJECTIVE: The aim of this study was to identify the causal variants for GD in the region encompassing TSHR by a refining association study. DESIGN AND METHODS: GD patients (1536) and 1516 sex-matched controls were recruited in the first stage, and an additional 3832 GD patients and 3426 sex-matched controls were recruited in the replication stage. Genotyping was performed using Illumina Human660-Quad BeadChips or TaqMan single nucleotide polymorphism (SNP) Genotyping Assays and the Fluidigm EP1 platform. RESULTS: When the results of regression analysis for 74 genotyped SNPs and 922 imputed SNPs in the first-stage cohort were combined, rs179243 and rs3783949 were the probable susceptibility SNPs associated with GD in TSHR. Eleven SNPs, including rs179243 and rs3783949, were selected to further refine the association in the replication study. Finally, rs12101261 and rs179243 were confirmed as independent GD susceptibility variants in the replication and combined populations. Further, we also found that the rate of persistent TSHR autoantibody positivity (pTRAb+) was significantly higher in the GD patients with the susceptible genotypes rs12101261 or rs179243 than in the GD patients carrying the protective genotypes, after the GD patients had been treated for more than 1 year. CONCLUSIONS: These findings indicate that rs12101261 and rs179243 are the possible causal SNPs for GD susceptibility in the TSHR gene and could serve as genetic markers to predict the outcome of pTRAb+ in GD patients.


Subject(s)
Genetic Predisposition to Disease , Graves Disease/genetics , Polymorphism, Single Nucleotide , Receptors, Thyrotropin/genetics , Antithyroid Agents/therapeutic use , Asian People , Case-Control Studies , China , Cohort Studies , Combined Modality Therapy , Drug Resistance , Female , Genetic Association Studies , Genetic Loci , Graves Disease/immunology , Graves Disease/metabolism , Graves Disease/therapy , Humans , Immunoglobulins, Thyroid-Stimulating/analysis , Iodine Radioisotopes/therapeutic use , Male , Radiopharmaceuticals/therapeutic use , Receptors, Thyrotropin/antagonists & inhibitors , Receptors, Thyrotropin/metabolism , Reproducibility of Results
19.
Hum Genet ; 133(5): 661-71, 2014 May.
Article in English | MEDLINE | ID: mdl-24346624

ABSTRACT

The BACH2 gene regulates B cell differentiation and function and has been reported to be a shared susceptibility gene for several autoimmune diseases. Our previous genome-wide association study (GWAS) indicated that several single nucleotide polymorphisms (SNPs) in the BACH2 gene are associated with Graves' disease (GD) in the Chinese Han population; however, the association did not achieve genome-wide significance levels. Recently, this association of BACH2 with GD was confirmed in Caucasians in the UK population, but fine mapping in this region has not yet been reported. Here, we provide a refined analysis of a 331-kb region in the BACH2 gene, which harbors 359 SNPs, using GWAS data from 1,442 GD patients and 1,468 controls. The SNPs rs2474619 and rs9344996 were implied as the independent variants associated with GD by forward and two-locus logistic regression analysis. We genotyped eight out of 10 tagSNPs with P < 1 × 10(-3) in 3,508 GD patients and 3,209 controls, the results also showed that rs2474619 was independently associated with GD in the combined population from GWAS and the second stage (P = 1.81 × 10(-5)). The rs2474619 and rs9344996 were further genotyped in the third stage cohorts, and rs2474619 showed evidence of association with GD at genome-wide significance levels in the combined population (P = 3.28 × 10(-8), odds ratio = 1.13). The association of rs9344996 with GD can be explained by its linkage to rs2474619 in the combined population. Our study clearly demonstrated that BACH2 is a susceptibility gene for GD in the Chinese Han population and further supported rs2474619, in intron 2 of BACH2, is the best association signal with GD. However, the mechanism by which BACH2 confers increased risk of GD requires further study.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Ethnicity/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Graves Disease/genetics , Autoimmune Diseases/genetics , Base Sequence , China , DNA Primers , Humans , Logistic Models , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction
20.
Hum Mol Genet ; 22(16): 3347-62, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23612905

ABSTRACT

Graves' disease (GD), characterized by autoantibodies targeting antigens specifically expressed in thyroid tissues causing hyperthyroidism, is triggered by a combination of genetic and environmental factors. However, only a few loci for GD risk were confirmed in the various ethnic groups, and additional genetic determinants have to be detected. In this study, we carried out a three-stage study in 9529 patients with GD and 9984 controls to identify new risk loci for GD and found genome-wide significant associations in the overall populations for five novel susceptibility loci: the GPR174-ITM2A at Xq21.1, C1QTNF6-RAC2 at 22q12.3-13.1, SLAMF6 at 1q23.2, ABO at 9q34.2 and an intergenic region harboring two non-coding RNAs at 14q32.2 and one previous indefinite locus, TG at 8q24.22 (Pcombined < 5 × 10(-8)). The genotypes of corresponding variants at 14q32.2 and 8q24.22 were correlated with the expression levels of C14orf64 and a TG transcript skipping exon 46, respectively. This study increased the number of GD loci with compelling evidence and indicated that non-coding RNAs might be potentially involved in the pathogenesis of GD.


Subject(s)
Genetic Predisposition to Disease , Graves Disease/genetics , RNA, Untranslated/genetics , Tumor Necrosis Factors/genetics , ABO Blood-Group System/genetics , Adult , Antigens, CD/genetics , Base Sequence , Case-Control Studies , Collagen , DNA, Intergenic , Female , Genetic Loci , Genome-Wide Association Study , Genotype , Humans , Male , Membrane Proteins/genetics , Middle Aged , Molecular Sequence Data , Polymorphism, Single Nucleotide , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/genetics , Signaling Lymphocytic Activation Molecule Family , Signaling Lymphocytic Activation Molecule Family Member 1
SELECTION OF CITATIONS
SEARCH DETAIL
...