Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
HLA ; 103(2): e15351, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38318996

ABSTRACT

HLA-B*51:389 differs from HLA-B*51:02:01:01 by one nucleotide in exon 2.


Subject(s)
HLA-B Antigens , Nucleotides , Humans , Alleles , Sequence Analysis, DNA , HLA-B Antigens/genetics , China
2.
Psychiatry Res ; 328: 115453, 2023 10.
Article in English | MEDLINE | ID: mdl-37660582

ABSTRACT

Clinical and etiological variability of attention deficit hyperactivity disorder (ADHD) presents an obstacle to understand the disorder. The aim of this study was to disentangle the heterogeneity of ADHD using neuroimaging and a semi-supervised machine learning algorithm. We collected brain structural and functional magnetic resonance imaging (MRI) data and clinical profiles of 183 children with ADHD and 396 neurotypical controls from 7 independent sites. We also used an external validation set with 750 subjects. We adopted a semi-supervised clustering method to subtype ADHD by regional volumetric measures of gray matter, white matter, and fractional amplitude of low frequency fluctuation (fALFF). In addition, split sample test, leave-one-site-out test and external validation were applied to evaluate the reproducibility and stability of ADHD subtypes. Two stable and reproducible neurosubtypes of ADHD were disclosed, which were proved by the split-sample test and leave-one-site-out validation. The structural and functional patterns of ADHD subtypes were also stable in the external validation set. The current two neurosubtypes differed in clinical manifestations and volumetric gray matter, white matter volume and fALFF patterns. The current neurosubtypes of ADHD which were different from clinical phenotypes could facilitate understanding the underlying neuropathological and neurobiological mechanism of the disorder.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Child , Humans , Attention Deficit Disorder with Hyperactivity/diagnostic imaging , Reproducibility of Results , Brain , Gray Matter/diagnostic imaging , Gray Matter/pathology , Magnetic Resonance Imaging/methods , Phenotype
3.
Ther Adv Med Oncol ; 15: 17588359231174869, 2023.
Article in English | MEDLINE | ID: mdl-37333902

ABSTRACT

Background: Only a fraction of patients with esophageal squamous cell carcinoma (ESCC) show tumor responses to anti-programmed cell death protein 1 (PD-1) therapy. The predictive value of single biomarkers for prognosis is limited, and a more comprehensive approach that incorporates multiple factors may improve the prognostic prediction. Here, we conducted a retrospective study to develop a combined immune prognostic index (CIPI) for predicting clinical outcomes of ESCC patients treated with anti-PD-1 therapy. Design and methods: We performed a pooled analysis of two multicenter clinical trials comparing immunotherapy versus chemotherapy as second-line treatment in ESCC patients. The discovery cohort comprised patients who received anti-PD-1 inhibitors (N = 322) and the control cohort comprised patients who received chemotherapy (N = 307). The validation cohort included patients with pan-cancers treated with PD-1/programmed cell death ligand-1 inhibitors, except for ESCC (N = 110). Multivariable Cox proportional hazard regression was used to assess the prediction value of variables on survival. Results: In the discovery cohort, neutrophil-to-lymphocyte ratio, serum albumin, and liver metastasis were independently associated with overall survival (OS) and progression-free survival (PFS). We integrated the three variables into CIPI and found that CIPI could categorize patients into four subgroups (CIPI 0 to CIPI 3) with distinct OS, PFS, and tumor responses. The CIPI was also predictive of clinical outcomes in the validation cohort, but not in the control cohort. Furthermore, patients with CIPI 0, CIPI 1, and CIPI 2 were more likely to benefit from anti-PD-1 monotherapy than chemotherapy, while patients with CIPI 3 did not benefit from anti-PD-1 monotherapy over chemotherapy. Conclusions: The CIPI score was a robust biomarker for prognostic prediction in ESCC patients treated with anti-PD-1 therapy and was immunotherapy specific. The CIPI score may also be applicable for prognostic prediction in pan-cancers.

4.
Ann Transl Med ; 11(5): 199, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-37007564

ABSTRACT

Background: Although immunotherapy combined with targeted therapy can be effective for hepatocellular carcinoma (HCC), not all HCC patients respond to this treatment. Models for predicting tumour response in HCC patients receiving immunotherapy combined with targeted therapy are lacking. Methods: A total of 221 HCC patients from two independent prospective cohorts were retrospectively reviewed. The patients were randomly divided into training and validation cohorts at a ratio of 7:3. Standard clinical data were collected from each patient, including age, sex, hepatitis B infection status, laboratory tests, and immune target-related adverse events (itrAEs). Tumour responses were evaluated using the Response Evaluation Criteria in Solid Tumours (RECIST) v1.1 guidelines. ItrAEs were assessed based on the Common Terminology Criteria for Adverse Events version 4.0. The nomogram for tumour response prediction was constructed based on the results of the multivariate logistic regression analysis, areas under the receiver operating characteristic curves (AUROCs) were used to determine the sensitivity and specificity of the model, and calibration plots and Hosmer-Lemeshow chi-square tests were performed to assess the calibration of the model. Results: In the multivariate logistic regression analysis, a solitary tumour (P=0.006), neutropenia (P=0.003) and hypertension (P=0.042) independently predicted objective response (OR). A nomogram for OR was established with AUROCs of 0.734, 0.675, 0.730, and 0.707 in the training, validation, first-line and second-line treatment sets, respectively. Tumour sizes less than 5 cm (P=0.005), a solitary tumour (P=0.037), prognostic nutritional indices greater than or equal to 54.3 (P=0.037), neutropenia (P=0.004) and fatigue (P=0.041) independently predicted disease control (DC). A nomogram for DC was established with AUROCs of 0.804, 0.667, and 0.768 in the training, first-line and second-line treatment sets, respectively. All the Hosmer-Lemeshow tests and calibration curves showed acceptable calibration. Conclusions: The current provides clinicians with new insights into selecting patients for immunotherapy combined with targeted therapy and contributes to the development of immunotherapy for HCC. It is necessary to expand the scale of our research and perform prospective studies to verify our findings.

5.
Oncologist ; 27(8): e625-e632, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35647908

ABSTRACT

BACKGROUND: Neuroendocrine neoplasms (NENs) are a group of diseases that show high heterogeneity but have limited treatment options. This phase I study evaluated the safety and efficacy of sintilimab, anti-PD-1 monoclonal antibody, in treating advanced NENs. METHODS: We prospectively enrolled patients pathologically diagnosed with NENs after standard treatment failure. Neuroendocrine neoplasms were classified into well-differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine cancers (NECs). Every patient received sintilimab, and response was assessed every 9 weeks. RESULTS: Twenty-four patients with a median age of 57.0 years were enrolled from November 2016 to 2017. The median Ki-67 index was 60%. Five patients had NET, 1 had NET G3, 17 had NEC, and 1 had mixed adenocarcinoma-neuroendocrine carcinoma. The most common primary tumor sites were the pancreas and gastrointestinal tract in 7 and 10 patients, respectively. In phase Ia trial, 2 patients received sintilimab 1 mg/kg every 2 weeks, one received 3 mg/kg every 2 weeks, and 21 patients enrolled in the phase Ib trial received 200 mg every 3 weeks. The objective response rate was 20.8% in all enrolled patients and 27.8% in NEC patients. The median progression-free survival was 2.2 and 2.1 months in patients with NET and NEC, respectively. The median OS was not applicable (NA) and 10.8 months (95% CI, 4.3, NA) with NET and NEC, respectively. The duration of response (DOR) was not reached, with a median follow-up time of 20.7 months. Treatment-related adverse events (TRAE) occurred in 17 (70.8%) patients. The most frequent TRAE was thyroid dysfunction (41.7%), and a grade 3 pulmonary infection occurred in 1 patient. The programmed cell death 1-ligand 1 (PD-L1)-positive (tumor proportion score ≥1%) rate was 18.8% (3 out of 16) and the expression of PD-L1 did not correlate with response. CONCLUSION: Sintilimab was well-tolerated and showed encouraging response in NECs. CLINICALTRIALS.GOV IDENTIFIER: NCT02937116.


Subject(s)
Carcinoma, Neuroendocrine , Neuroendocrine Tumors , Antibodies, Monoclonal, Humanized/adverse effects , B7-H1 Antigen , Carcinoma, Neuroendocrine/pathology , Humans , Middle Aged
6.
Cancer Immunol Immunother ; 71(6): 1443-1451, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34689233

ABSTRACT

BACKGROUND: Treatment strategies are limited for patients with chemotherapy refractory microsatellite stable (MSS) colorectal cancer. We aim to evaluate the efficacy and safety of immune checkpoint inhibitors (ICIs) combined with regorafenib in this population in routine clinical practice. METHODS: We retrospectively analyzed patients with advanced or metastatic colorectal cancer who received at least one dose of ICIs combined with regorafenib in 14 Chinese medical centers. The primary outcome was objective response rate (ORR). This study was registered at ClinicalTrials.gov on February 2020 (NCT04771715). RESULTS: Eighty-four patients received ICIs combined with regorafenib from January 2019 to January 2021. Most patients (91%) received two or more systemic treatment lines before the study treatment. Seventy-six patients (90%) had confirmed MSS status. At a median follow-up of 5.5 months, four patients achieved partial response (5%) and 37 patients achieved stable disease (45%) as the best response. The median progression-free survival (PFS) was 3.1 months, and the median overall survival was 17.3 months. Eleven patients (13%) remained progression-free for more than 6 months. Baseline liver metastasis (HR 1.98, 95%CI 1.07-3.69, P = 0.03) and neutrophil-lymphocyte ratio (NLR) of ≥ 1.5 (HR 2.83, 95%CI 1.00-7.98, P = 0.05) were associated with shorter PFS in multivariate analysis. Grade 3 or higher treatment-related adverse events (TRAEs) occurred in 16 patients (19%). CONCLUSION: The combination of ICIs with regorafenib can be a valuable treatment option for a proportion of patients with chemotherapy refractory MSS colorectal cancer. Patients with no liver metastasis and a low NLR at baseline may derive most benefit from this strategy.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Liver Neoplasms/drug therapy , Microsatellite Repeats , Phenylurea Compounds/therapeutic use , Pyridines , Retrospective Studies
7.
BMC Gastroenterol ; 21(1): 398, 2021 Oct 23.
Article in English | MEDLINE | ID: mdl-34688250

ABSTRACT

BACKGROUND: Larotinib is a new first-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor. This open-label, phase 1b study is aimed to evaluate the efficacy, safety of larotinib in patients with advanced esophageal squamous cell carcinoma (ESCC) with EGFR overexpression or amplification pretreated with one or more system regimens, and to recommend an appropriate dose for its further study. METHODS: Patients received larotinib orally at 3 doses (250, 300, 350 mg), once daily. Clinical response was evaluated every 8 weeks according to RECIST v1.1 criteria by both investigators and independent radiology review (IRC). RESULTS: 81 patients were enrolled. The investigator-assessed overall response rate (ORR) was 13.7% (10/73), all responses were observed in the 350 mg group of which ORR up to 20.0% (10/50), with 10 of them having EGFR overexpression and 4 having EGFR amplification. Per IRC assessment, ORR for all patients and 350 mg group were 13.9% (10/72) and 16.3% (8/50). In the 350 mg group, median overall survival (OS) and progression-free survival (PFS) were 8.0 (95% CI 4.9-10.2) months and 3.4 (95% CI 2.4-3.7) months, respectively. The most common treatment-related adverse events (TRAEs) were diarrhea, rash, and palmar-plantar erythrodysesthesia syndrome, elevated AST/ALT, vomiting, similarly with other EGFR TKIs. CONCLUSIONS: Larotinib demonstrated promising antitumor activity and manageable safety profiles in patients with pre-treated advanced ESCC with EGFR overexpression or amplification, especially at the dose of 350 mg, which showed better efficacy and acceptable safety. A phase 3 study is underway on 350 mg larotinib in ESCC patients with EGFR overexpression. TRIAL REGISTRATION: This trial was retrospectively registered on 25/03/2019, NCT03888092. https://clinicaltrials.gov/ct2/show/NCT03888092 .


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Lung Neoplasms , Antineoplastic Agents/adverse effects , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/genetics , ErbB Receptors/therapeutic use , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Humans , Lung Neoplasms/drug therapy , Mutation , Protein Kinase Inhibitors/adverse effects
8.
Oncologist ; 26(12): e2227-e2238, 2021 12.
Article in English | MEDLINE | ID: mdl-34516729

ABSTRACT

BACKGROUND: Programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) blockade immunotherapies have changed the landscape of cancer therapy. However, the main limitation of these therapies is the lack of definitively predictive biomarkers to predict treatment response. Whether PD-L1 expression on circulating tumor cells (CTCs) is associated with the clinical outcomes of immunotherapy remains to be extensively investigated. MATERIALS AND METHODS: One hundred fifty-five patients with different advanced cancers were enrolled in this study and treated with anti-PD-1/PD-L1 monoclonal antibodies. Using the Pep@MNPs method, CTCs were isolated and enumerated. The PD-L1 expression levels were analyzed by an immunofluorescence assay for semiquantitative assessment with four categories (negative, low, medium, and high). RESULTS: Prior to immunotherapy, 81.93% (127/155) of patients had PD-L1-positive CTCs, and 71.61% (111/155) had at least one PD-L1-high CTC. The group with PD-L1-positive CTCs had a higher disease control rate (DCR) (71.56%, 91/127), with a DCR of only 39.29% (11/28) for the remaining individuals (p = .001). The objective response rate and DCR in PD-L1-high patients were higher than those in the other patients (32.44% vs. 13.64%, p = .018 and 75.68% vs. 40.91%, p < .0001, respectively). The reduction in the counts and ratios of PD-L1-positive CTCs and PD-L1-high CTCs reflected a beneficial response to PD-1/PD-L1 inhibitors. Furthermore, patients with PD-L1-high CTCs had significantly longer progression-free survival (4.9 vs. 2.2 months, p < .0001) and overall survival (16.1 vs. 9.0 months, p = .0235) than those without PD-L1-high CTCs. CONCLUSION: The PD-L1 level on CTCs may serve as a clinically actionable biomarker for immunotherapy, and its dynamic changes could predict the therapeutic response. IMPLICATIONS FOR PRACTICE: This study was designed to investigate the role of programmed death-ligand 1 (PD-L1) expression on circulating tumor cells in predicting and monitoring response to programmed death-1 (PD-1)/PD-L1 blockade immunotherapies in patients with advanced cancer. The results of the study showed that PD-L1-high-expression circulating tumor cells (CTCs) were both a predictive biomarker and a prognostic factor in patients with advanced cancer treated with anti-PD-1/PD-L1 monoclonal antibodies. These observations suggest that PD-L1 level on CTCs is a potential clinical biomarker for immunotherapy.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Neoplasms , Neoplastic Cells, Circulating , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Humans , Immunotherapy , Neoplasms/diagnosis , Neoplasms/drug therapy
9.
Cancer Immunol Immunother ; 70(9): 2669-2679, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33624146

ABSTRACT

Immune checkpoint blockade (ICB) of the programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) immune checkpoint pathway has led to unprecedented advances in cancer therapy. However, the overall response rate of anti-PD-1/PD-L1 monotherapy is still unpromising, underscoring the need for predictive biomarkers. In this retrospective study, we collected pretreatment plasma samples from two independent cohorts of patients receiving ICB. To determine whether a signature of plasma cytokines could be associated with therapeutic efficacy, we systemically profiled cytokine clusters and functional groups in the discovery and validation datasets by using 59 multiplexed bead immunoassays and bioinformatics analysis. We first attempted to functionally classify the 59 immunological factors according to their biological classification or functional roles in the cancer-immunity cycle. Surprisingly, we observed that two signatures, the "checkpoint signature" and "trafficking of T-cell signature", were higher in the response subgroup than in the nonresponse subgroup in both the discovery and validation cohorts. Moreover, enrichment of the "checkpoint signature" was correlated with improved overall survival and progression-free survival in both datasets. In addition, we demonstrated that increased baseline levels of three checkpoint molecules (PD-L1, T-cell immunoglobulin mucin receptor 3 and T-cell-specific surface glycoprotein CD28) were common peripheral responsive correlates in both cohorts, thus rendering this "refined checkpoint signature" an ideal candidate for future verification. In the peripheral blood system, the "refined checkpoint signature" may function as a potential biomarker for anti-PD-1/PD-L1 monotherapy in gastrointestinal (GI) cancers.


Subject(s)
Biomarkers, Tumor , Cytokines/metabolism , Gastrointestinal Neoplasms/etiology , Immune Checkpoint Proteins/metabolism , Computational Biology , Cytokines/genetics , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/metabolism , Gene Expression Profiling , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Proteins/genetics , Immunohistochemistry , Kaplan-Meier Estimate , Male , Prognosis , Reproducibility of Results , Treatment Outcome
10.
Oncol Lett ; 21(1): 68, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33365079

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is a highly malignant and deadly tumor. Radiation therapy is one of the primary treatments for locally advanced ESCC. However, the biomarkers for prognosis of definitive radiation remain undefined. Peripheral blood circulating tumor (ct)DNA provides information of tumor genetic alterations and has been confirmed as a potential non-invasive biomarker for several types of cancer. The present study investigated the clinical implications of ctDNA detection in patients with ESCC and receiving definitive radiation therapy. Patients with locally advanced ESCC were retrospectively recruited. Plasma samples were collected before, during and following radiation therapy. Next-generation sequencing was performed to identify somatic mutations in 180 genes. A total of 69 baseline and post-radiation plasma samples were collected from 25 patients. A total of 59 non-silent single nucleotide variants were present in 33 genes. All pre-radiation and 58.3% (14/24) of post-radiation samples had at least one mutation. Patients with lymph node metastases (LNM) exhibited a higher number of pre-radiation mutations compared with those without LNM. The variables, progression-free survival (PFS) and overall survival (OS) of the patients with one baseline mutation were not significantly different compared with that in patients with more than one baseline mutation. Patients with initial ctDNA-positive post-radiation samples exhibited significantly reduced PFS (P=0.047) and OS (P=0.005) compared with that in patients with ctDNA-negative samples. The post-radiation plasma ctDNA status was an independent prognostic factor from univariate and multivariate analyses. Dynamic monitoring of ctDNA during follow-up was examined. The results indicated that ctDNA was a predictive and prognostic marker in patients with ESCC and receiving definitive radiation therapy, which may guide subsequent treatment.

11.
Chin J Cancer Res ; 32(3): 370-382, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32694901

ABSTRACT

OBJECTIVE: Fluzoparib (SHR3162) is a novel, potent poly(ADP-ribose) polymerases (PARP)1, 2 inhibitor that showed anti-tumor activity in xenograft models. We conducted a phase I, first-in-human, dose-escalation and expansion (D-Esc and D-Ex) trial in patients with advanced solid cancer. METHODS: This was a 3+3 phase I D-Esc trial with a 3-level D-Ex at 5 hospitals in China. Eligible patients for D-Esc had advanced solid tumors refractory to standard therapies, and D-Ex enrolled patients with ovarian cancer (OC). Fluzoparib was administered orally once or twice daily (bid) at 11 dose levels from 10 to 400 mg/d. Endpoints included dose-finding, safety, pharmacokinetics, and antitumor activity. RESULTS: Seventy-nine patients were enrolled from March, 2015 to January, 2018 [OC (47, 59.5%); breast cancer (BC) (16, 20.3%); colorectal cancer (8, 10.1%), other tumors (8, 10.1%)]; 48 patients were treated in the D-Esc arm and 31 in the D-Ex arm. The maximum tolerated dose (MTD) was 150 mg bid, with a half-life of 9.14 h. Grade 3/4 adverse events included anemia (7.6%) and neutropenia (5.1%). The objective response rate (ORR) was 30% (3/10) in patients with platinum-sensitive OC and 7.7% (1/13) in patients with BC. Among patients treated with fluzoparib ≥120 mg/d, median progression-free survival (mPFS) was 7.2 [95% confidence interval (95% CI), 1.8-9.3] months in OC, 9.3 (95% CI, 7.2-9.3) months in platinum-sensitive OC, and 3.5 (range, 2.0-28.0) months in BC. In patients with germline BC susceptibility gene mutation (gBRCA Mut) (11/43 OC; 2/16 BC), mPFS was 8.9 months for OC (range, 1.0-23.2; 95% CI, 1.0-16.8) and 14 and 28 months for BC (those two patients both also had somaticBRCA Mut). CONCLUSIONS: The MTD of fluzoparib was 150 mg bid in advanced solid malignancies. Fluzoparib demonstrated single-agent antitumor activity in BC and OC, particularly in BRCA Mut and platinum-sensitive OC.

12.
Sci Adv ; 6(1): eaax5576, 2020 01.
Article in English | MEDLINE | ID: mdl-31911942

ABSTRACT

Cetuximab improves the survival of patients with metastatic colorectal cancer. The main limitation is primary and secondary resistance, the underlying mechanism of which requires extensive investigation. We proved that PRSS expression levels are significantly negatively associated with the sensitivity of cancer cells to cetuximab. Detailed mechanistic analysis indicated that PRSS can cleave cetuximab, leading to resistance. Cetuximab or bevacizumab combined with SPINK1, a PRSS inhibitor, inhibited cell growth more efficiently than cetuximab or bevacizumab alone in xenograft models. PRSS levels in the serum of 156 patients with mCRC were analyzed, and poor efficacy of cetuximab therapy was observed in patients with aberrant PRSS expression. PRSS expression in monoclonal antibody (mAb)-treated patients with cancer from The Cancer Genome Atlas database was also evaluated to determine whether patients with higher PRSS expression have significantly reduced progression-free survival. Our work provides a strong scientific rationale for targeting PRSS in combination with cetuximab therapy.


Subject(s)
Cetuximab/administration & dosage , Colorectal Neoplasms/drug therapy , Trypsin Inhibitor, Kazal Pancreatic/genetics , Trypsin/genetics , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab , Cell Line, Tumor , Cetuximab/adverse effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease-Free Survival , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Male , Mice , Middle Aged , Neoplasm Metastasis
13.
EBioMedicine ; 43: 261-269, 2019 May.
Article in English | MEDLINE | ID: mdl-31031019

ABSTRACT

BACKGROUND: Circulating tumor DNA (ctDNA) isolated from plasma contains genetic mutations that can be representative of those found in primary tumor tissue DNA. These samples can provide insights into tumoral heterogeneity in patients with advanced gastric cancer (AGC). Although trastuzumab has been shown to be effective in first-line therapy for patients with metastatic gastric cancer with overexpression of human epidermal growth factor receptor 2 (HER2), the mechanism of AGC resistance is incompletely understood. METHODS: In this prospective study, we used targeted capture sequencing to analyze 173 serial ctDNA samples from 39 AGC patients. We analyzed cancer cell fractions with PyClone to understand the clonal population structure in cancer, and monitored serial samples during therapy. Serial monitoring of ctDNA using the molecular tumor burden index (mTBI), identified progressive disease before imaging results (mean: 18 weeks). FINDINGS: We reconstructed the clonal structure of ctDNA during anti-HER2 treatment, and identified 32 expanding mutations potentially related to trastuzumab resistance. Multiple pathways activating in the same patients revealed heterogeneity in trastuzumab resistance mechanisms in AGC. In patients who received chemotherapy, mTBI was validated for the prediction of progressive disease, with a sensitivity of 94% (15/16). A higher mTBI (≥1%) in pretreatment ctDNA was also a risk factor for progression-free survival. CONCLUSIONS: Analysis of ctDNA clones based on sequencing is a promising approach to clinical management, and may lead to improved therapeutic strategies for AGC patients. FUND: This work was supported by grants from the National International Cooperation Grant (to J.X.; Project No. 2014DFB33160).


Subject(s)
Biomarkers, Tumor , Circulating Tumor DNA , Drug Resistance, Neoplasm/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Adult , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Disease Progression , Female , Humans , Male , Middle Aged , Molecular Targeted Therapy , Mutation , Neoplasm Grading , Neoplasm Staging , Prognosis , Proportional Hazards Models , Receptor, ErbB-2/antagonists & inhibitors , Stomach Neoplasms/blood , Stomach Neoplasms/drug therapy , Tomography, X-Ray Computed , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Treatment Outcome
14.
Nat Commun ; 10(1): 39, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604760

ABSTRACT

The human gastric mucosa is the most active layer of the stomach wall, involved in food digestion, metabolic processes and gastric carcinogenesis. Anatomically, the human stomach is divided into seven regions, but the protein basis for cellular specialization is not well understood. Here we present a global analysis of protein profiles of 82 apparently normal mucosa samples obtained from living individuals by endoscopic stomach biopsy. We identify 6,258 high-confidence proteins and estimate the ranges of protein expression in the seven stomach regions, presenting a region-resolved proteome reference map of the near normal, human stomach. Furthermore, we measure mucosa protein profiles of tumor and tumor nearby tissues (TNT) from 58 gastric cancer patients, enabling comparisons between tumor, TNT, and normal tissue. These datasets provide a rich resource for the gastrointestinal tract research community to investigate the molecular basis for region-specific functions in mucosa physiology and pathology including gastric cancer.


Subject(s)
Gastric Mucosa/metabolism , Neoplasm Proteins/analysis , Proteome/analysis , Stomach Neoplasms/pathology , Biopsy , Carcinogenesis/pathology , Cardia/metabolism , Cardia/pathology , Datasets as Topic , Gastric Fundus/metabolism , Gastric Fundus/pathology , Gastric Mucosa/pathology , Gastroscopy , Humans , Neoplasm Proteins/metabolism , Proteome/metabolism , Proteomics/methods , Pyloric Antrum/metabolism , Pyloric Antrum/pathology , Pylorus/metabolism , Pylorus/pathology
15.
Clin Cancer Res ; 25(2): 515-523, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30348638

ABSTRACT

PURPOSE: This study assessed the safety and efficacy of SHR-1210 (anti-PD-1 antibody) and apatinib (VEGFR2 inhibitor) as combination therapy in patients with advanced hepatocellular carcinoma (HCC), gastric, or esophagogastric junction cancer (GC/EGJC). PATIENTS AND METHODS: This was an open-label, dose-escalation (phase Ia) and expansion study (phase Ib). In phase Ia, patients (n = 15) received SHR-1210 200 mg every 2 weeks and apatinib 125-500 mg once daily until unacceptable toxicity or disease progression. In phase Ib, patients (n = 28) received apatinib at the phase Ia-identified recommended phase II dose (RP2D) plus SHR-1210. The primary objectives were safety and tolerability and RP2D determination. RESULTS: At data cutoff, 43 patients were enrolled. In phase Ia, four dose-limiting toxicity events were observed (26.7%): one grade 3 lipase elevation (6.7%) in the apatinib 250 mg cohort and three grade 3 pneumonitis events (20%) in the apatinib 500 mg cohort. The maximum tolerated RP2D for apatinib was 250 mg. Of the 33 patients treated with the R2PD combination, 20 (60.6%) experienced a grade ≥3 treatment-related adverse event; adverse events in ≥10% of patients were hypertension (15.2%) and increased aspartate aminotransferase (15.2%). The objective response rate in 39 evaluable patients was 30.8% (95% CI: 17.0%-47.6%). Eight of 16 evaluable HCC patients achieved a partial response (50.0%, 95% CI: 24.7%-75.4%). CONCLUSIONS: SHR-1210 and apatinib combination therapy demonstrated manageable toxicity in patients with HCC and GC/EGJC at recommended single-agent doses of both drugs. The RP2D for apatinib as combination therapy was 250 mg, which showed encouraging clinical activity in patients with advanced HCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Esophageal Neoplasms/drug therapy , Esophagogastric Junction/pathology , Liver Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/mortality , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/etiology , Female , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/etiology , Liver Neoplasms/mortality , Male , Middle Aged , Molecular Targeted Therapy , Prognosis , Pyridines/administration & dosage , Stomach Neoplasms/diagnosis , Stomach Neoplasms/etiology , Stomach Neoplasms/mortality , Treatment Outcome
16.
Blood Rev ; 32(3): 192-202, 2018 05.
Article in English | MEDLINE | ID: mdl-29174416

ABSTRACT

Acute leukemia is a global disease with a poor prognosis for many patients. While an increasing number of patients with acute leukemia are being treated with unrelated hematopoietic stem cell transplants (HSCT) or umbilical cord blood transplants (UCBT), recent comparative reports of these 2 procedures are lacking. Therefore, we conducted a meta-analysis of the safety and efficacy of unrelated HSCT and unrelated single-unit UCBT for the treatment of pediatric and adult patients with acute lymphoblastic leukemia (ALL) or acute myeloid leukemia (AML). The primary outcomes were the rates of relapse, overall survival (OS), progression-free survival (PFS), and days to neutrophil and platelet recovery. Pooled effects (odds ratios [OR] or difference in means) were determined with either random- or fixed-effects models. Our meta-analysis included 9 studies that enrolled 6762 patients (UCBT: n=2026; HSCT: n=4736). The risk of relapse for patients undergoing UCBT was similar to that of patients who received an HSCT (OR=1.030; 95%CI, 0.767 to 1.383, P=0.847). We also found no difference between HSCT and UCBT for OS (pooled OR=1.417; 95%CI, 0.936 to 2.146; P=0.100) or PFS (OR=1.165; 95%CI, 0.996 to 1.362; P=0.056). However, neutrophil and platelet recovery periods were both shorter after HSCT than after UCBT (neutrophil recovery: difference in means=-3.420, 95% CI: -5.491 to -1.349, P=0.001; platelet recovery: difference in means -20.350, 95% CI: -33.656 to -7.044, P=0.003). Collectively, our data provide strong evidence to support increased use of cord blood transplants for both adults and children with acute leukemia.


Subject(s)
Cord Blood Stem Cell Transplantation , Leukemia, Myeloid, Acute/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Unrelated Donors , Cord Blood Stem Cell Transplantation/adverse effects , Cord Blood Stem Cell Transplantation/methods , Graft vs Host Disease/etiology , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Leukocyte Count , Neutrophils , Odds Ratio , Peripheral Blood Stem Cell Transplantation/adverse effects , Peripheral Blood Stem Cell Transplantation/methods , Platelet Count , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Recurrence , Treatment Outcome
17.
Exp Ther Med ; 14(2): 1427-1432, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28810606

ABSTRACT

The present case study reported on a 62-year-old male patient with gastric cancer-associated Aspergillus (A.) niger bloodstream infection. The patient presented with massive hemorrhage in the gastrointestinal tract 3 months after total gastrectomy for gastric cancer. Conservative treatment consisting of blood transfusion to supplement blood volume loss was ineffective. Digital subtraction angiography indicated gastroduodenal artery bleeding. The first attempt of performing arterial embolization using gelatin sponges failed, while the second attempt of performing common hepatic artery embolization using gelatin sponges and micro-coil springs stopped the bleeding. Four weeks after angiography, the patient presented with the complication of A. niger bloodstream infection, which was cured using intravenous and oral voriconazole. Clinicians should be aware of the possibility of A. niger bloodstream infection after invasive operations in immunocompromised patients and apply timely antifungal treatment.

18.
Clin Cancer Res ; 23(16): 4602-4616, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28424201

ABSTRACT

Purpose: Mutations in KRAS are considered to be the main drivers of acquired resistance to epidermal growth factor receptor (EGFR) blockade in patients with metastatic colorectal cancer (mCRC). However, the potential role of other genes downstream of the EGFR signaling pathway in conferring acquired resistance has not been extensively investigated.Experimental Design: Using circulating tumor DNA (ctDNA) from patients with mCRC and with acquired cetuximab resistance, we developed a targeted amplicon ultra-deep sequencing method to screen for low-abundance somatic mutations in a panel of genes that encode components of the EGFR signaling pathway. Mutations with significantly increased variant frequencies upon disease progression were selected by using quartile analysis. The functional consequences of the identified mutations were validated in cultured cells.Results: We analyzed 32 patients with acquired cetuximab resistance in a development cohort. Of them, seven (22%) carried five novel PIK3CA mutations, whereas eight (25%) carried previously reported KRAS mutations. Functional studies showed that novel PIK3CA mutations (all in exon 19; p.K944N, p.F930S, p.V955G, p.V955I, and p.K966E) promote cell viability in the presence of cetuximab. Only one novel PIK3CA mutation (p.K944N) was verified in one of the 27 patients with acquired resistance in a validation cohort, simultaneous KRAS and PIK3CA hotspot mutations were detected in two patients. Among the above 59 acquired resistance patients, those with PIK3CA or RAS mutations detected in ctDNA showed a pronounced decrease in progression-free survival than patients with no mutation.Conclusions: The PIK3CA mutations may potentially contribute to acquired cetuximab resistance in patients with mCRC. Clin Cancer Res; 23(16); 4602-16. ©2017 AACR.


Subject(s)
Cetuximab/therapeutic use , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Mutation , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/therapeutic use , Circulating Tumor DNA/blood , Circulating Tumor DNA/chemistry , Circulating Tumor DNA/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Disease-Free Survival , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Proto-Oncogene Proteins p21(ras)/genetics , Retrospective Studies
19.
BMC Cancer ; 16(1): 753, 2016 Sep 23.
Article in English | MEDLINE | ID: mdl-27662833

ABSTRACT

BACKGROUND: Extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, can be secreted by most cell types and released in perhaps all biological fluids. EVs contain multiple proteins, specific lipids and several kinds of nucleic acids such as RNAs and DNAs. Studies have found that EVs contain double-stranded DNA and that genetic information has a certain degree of consistency with tumor DNA. Therefore, if genes that exist in exosomes are stable, we may be able to use EVs genetic testing as a new means to monitor gene mutation. METHODS: In this study, EVs were extracted from serum under various storage conditions (4 °C, room temperature and repeated freeze-thaw). We used western blotting to examine the stability of serum EVs. Then, we extracted DNA from EVs and tested the concentration changing under different conditions. We further assessed the stability of EVs DNA s using polymerase chain reaction (PCR) and Sanger sequencing. RESULTS: EVs is stable under the conditions of 4 °C (for 24 h, 72 h, 168 h), room temperature (for 6 h, 12 h, 24 h, 48 h) and repeated freeze-thaw (after one time, three times, five times). Also, serum DNA is mainly present in EVs, especially in exosomes, and that the content and function of DNA in EVs is stable whether in a changing environment or not. We showed that EVs DNA stayed stable for 1 week at 4 °C, 1 day at room temperature and after repeated freeze-thaw cycles (less than three times). However, DNA from serum EVs after 2 days at room temperature or after five repeated freeze-thaw cycles could be used for PCR and sequencing. CONCLUSIONS: Serum EVs and EVs DNA can remain stable under different environments, which is the premise that EVs could serve as a novel means for genetic tumor detection and potential biomarkers for cancer diagnostics and prognostics.

20.
Exp Ther Med ; 11(1): 201-207, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26889240

ABSTRACT

Primary lung enteric adenocarcinoma is a rare type of invasive lung carcinoma. Its morphology and immunohistochemistry are those of colorectal carcinoma, but there is no associated primary colorectal carcinoma. The present study describes the case of a 53-year-old female who presented with an irritating cough and a mass around the right sternoclavicular joint. Comprehensive evaluation revealed involvement of the mediastinum, lungs, right sternoclavicular joint and right kidney. Biopsies from the mediastinal and right sternoclavicular joint tumors showed features of adenocarcinoma. Immunohistochemistry was positive for cytokeratin (CK)20 and caudal type homeobox transcription factor 2, and negative for CK7, thyroid transcription factor-1 and napsin A. Genotypic analysis identified the expression of wild-type epidermal growth factor receptor, Kirsten rat sarcoma viral oncogene homolog, serine/threonine-protein kinase B-Raf and UDP-glucuronosyltransferase 1-1. There was no expression of echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase and a moderate expression of excision repair cross-complementation group 1, ribonucleoside-diphosphate reductase large subunit and tubulin ß-3 chain. A strong expression of thymidylate synthase and 677TC genotype expression of methylenetetrahydrofolate reductase was observed. Gastroscopy, enteroscopy, colorectal colonoscopy and positron emission tomography-computed tomography failed to find evidence of a gastrointestinal malignancy and primary lung enteric adenocarcinoma was diagnosed. The presence of multiple metastases did not permit curative surgery. The patient was treated with 3 monthly cycles of the XELOX chemotherapy regimen; the response was poor with progression of supraclavicular lesions. Treatment was switched to the TP regimen for 4 monthly cycles, which resulted in a significant reduction in the size of the lung lesions; however, the supraclavicular lesion responded poorly to the treatment. The patient then received 2 cycles of the FOLFIRI regimen; however, the lung and right supraclavicular lesions progressed, causing increased right upper limb pain. The pain was alleviated by palliative surgery. Following surgery, the DP regimen was employed. Follow-up of the patient remains ongoing. The present findings suggest that the early diagnosis and treatment of primary lung enteric adenocarcinoma is likely to improve patient outcome.

SELECTION OF CITATIONS
SEARCH DETAIL