Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Clin Invest ; 134(6)2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38271117

ABSTRACT

BACKGROUNDThe tumor immune microenvironment can provide prognostic and therapeutic information. We aimed to develop noninvasive imaging biomarkers from computed tomography (CT) for comprehensive evaluation of immune context and investigate their associations with prognosis and immunotherapy response in gastric cancer (GC).METHODSThis study involved 2,600 patients with GC from 9 independent cohorts. We developed and validated 2 CT imaging biomarkers (lymphoid radiomics score [LRS] and myeloid radiomics score [MRS]) for evaluating the IHC-derived lymphoid and myeloid immune context respectively, and integrated them into a combined imaging biomarker [LRS/MRS: low(-) or high(+)] with 4 radiomics immune subtypes: 1 (-/-), 2 (+/-), 3 (-/+), and 4 (+/+). We further evaluated the imaging biomarkers' predictive values on prognosis and immunotherapy response.RESULTSThe developed imaging biomarkers (LRS and MRS) had a high accuracy in predicting lymphoid (AUC range: 0.765-0.773) and myeloid (AUC range: 0.736-0.750) immune context. Further, similar to the IHC-derived immune context, 2 imaging biomarkers (HR range: 0.240-0.761 for LRS; 1.301-4.012 for MRS) and the combined biomarker were independent predictors for disease-free and overall survival in the training and all validation cohorts (all P < 0.05). Additionally, patients with high LRS or low MRS may benefit more from immunotherapy (P < 0.001). Further, a highly heterogeneous outcome on objective response ​rate was observed in 4 imaging subtypes: 1 (-/-) with 27.3%, 2 (+/-) with 53.3%, 3 (-/+) with 10.2%, and 4 (+/+) with 30.0% (P < 0.0001).CONCLUSIONThe noninvasive imaging biomarkers could accurately evaluate the immune context and provide information regarding prognosis and immunotherapy for GC.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/therapy , Radiomics , Immunotherapy , Tomography, X-Ray Computed , Tumor Microenvironment , Biomarkers , Prognosis
2.
Chem Commun (Camb) ; 60(7): 862-865, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38131618

ABSTRACT

One-pot synthesis of structurally diverse sulfurized/selenated 4-aminopyrimidines has been reported via t-BuOK/K2S2O8-promoted four-component reaction of mixed nitriles and disulfides/diselenides. Mechanistic studies indicate that the reaction proceeds through radical and ionic pathways, and an alkenyl sulfide serves as a key intermediate.

3.
BMJ Open ; 13(9): e071433, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37734891

ABSTRACT

OBJECTIVE: The primary objective of this study is to investigate the prevalence and risk factors of stroke in high-altitude areas through a comprehensive systematic review and meta-analysis. DESIGN: This study adopts a systematic review and meta-analysis design. DATA SOURCES: A thorough search was conducted on databases including PubMed, Web of Science, Embase, Cochrane Library, MEDLINE and SCOPUS, covering the period up to June 2023. ELIGIBILITY CRITERIA: Studies reporting the prevalence of stroke in high-altitude areas and exploring related risk factors were included, regardless of whether they involved clinical samples or the general population. Studies with incomplete, outdated or duplicate data were excluded. DATA EXTRACTION AND SYNTHESIS: We performed eligibility screening, data extraction and quality evaluation of the retrieved articles. Meta-analysis was employed to estimate the prevalence and risk factors of stroke in high-altitude areas. The Newcastle-Ottawa Scale was used to assess the risk of bias. RESULTS: A total of 17 studies encompassing 8 566 042 participants from four continents were included, with altitudes ranging from 1500 m to nearly 5000 m. The pooled prevalence of stroke in high-altitude areas was found to be 0.5% (95% CI 0.3%-7%). Notably, the prevalence was higher in clinical samples (1.2%; 0.4%-2.5%) compared with the general population (0.3%; 95% CI 0.1%-0.6%). When considering geographic regions, the aggregated data indicated that stroke prevalence in the Eurasia plate was 0.3% (0.2%-0.4%), while in the American region, it was 0.8% (0.4%-1.3%). Age (OR, 14.891), gender (OR, 1.289), hypertension (OR, 3.158) and obesity (OR, 1.502) were identified as significant risk factors for stroke in high-altitude areas. CONCLUSIONS: The findings of this study provide insights into the pooled prevalence of stroke in high-altitude areas, highlighting variations based on geographic regions and sampling type. Moreover, age, gender, hypertension and obesity were found to be associated with the occurrence of stroke. PROSPERO REGISTRATION NUMBER: CRD42022381541.


Subject(s)
Hypertension , Stroke , Humans , Altitude , Prevalence , Risk Factors , Obesity , Stroke/epidemiology , Stroke/etiology
4.
Nat Commun ; 14(1): 5135, 2023 08 23.
Article in English | MEDLINE | ID: mdl-37612313

ABSTRACT

Substantial progress has been made in using deep learning for cancer detection and diagnosis in medical images. Yet, there is limited success on prediction of treatment response and outcomes, which has important implications for personalized treatment strategies. A significant hurdle for clinical translation of current data-driven deep learning models is lack of interpretability, often attributable to a disconnect from the underlying pathobiology. Here, we present a biology-guided deep learning approach that enables simultaneous prediction of the tumor immune and stromal microenvironment status as well as treatment outcomes from medical images. We validate the model for predicting prognosis of gastric cancer and the benefit from adjuvant chemotherapy in a multi-center international study. Further, the model predicts response to immune checkpoint inhibitors and complements clinically approved biomarkers. Importantly, our model identifies a subset of mismatch repair-deficient tumors that are non-responsive to immunotherapy and may inform the selection of patients for combination treatments.


Subject(s)
Brain Neoplasms , Deep Learning , Humans , Immunotherapy , Chemotherapy, Adjuvant , Biology , Tumor Microenvironment
5.
Cell Rep Med ; 4(8): 101146, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37557177

ABSTRACT

The tumor microenvironment (TME) plays a critical role in disease progression and is a key determinant of therapeutic response in cancer patients. Here, we propose a noninvasive approach to predict the TME status from radiological images by combining radiomics and deep learning analyses. Using multi-institution cohorts of 2,686 patients with gastric cancer, we show that the radiological model accurately predicted the TME status and is an independent prognostic factor beyond clinicopathologic variables. The model further predicts the benefit from adjuvant chemotherapy for patients with localized disease. In patients treated with checkpoint blockade immunotherapy, the model predicts clinical response and further improves predictive accuracy when combined with existing biomarkers. Our approach enables noninvasive assessment of the TME, which opens the door for longitudinal monitoring and tracking response to cancer therapy. Given the routine use of radiologic imaging in oncology, our approach can be extended to many other solid tumor types.


Subject(s)
Deep Learning , Stomach Neoplasms , Humans , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/therapy , Tumor Microenvironment , Immunotherapy , Chemotherapy, Adjuvant
6.
Radiother Oncol ; 186: 109793, 2023 09.
Article in English | MEDLINE | ID: mdl-37414254

ABSTRACT

BACKGROUND AND PURPOSE: Immunotherapy is a standard treatment for many tumor types. However, only a small proportion of patients derive clinical benefit and reliable predictive biomarkers of immunotherapy response are lacking. Although deep learning has made substantial progress in improving cancer detection and diagnosis, there is limited success on the prediction of treatment response. Here, we aim to predict immunotherapy response of gastric cancer patients using routinely available clinical and image data. MATERIALS AND METHODS: We present a multi-modal deep learning radiomics approach to predict immunotherapy response using both clinical data and computed tomography images. The model was trained using 168 advanced gastric cancer patients treated with immunotherapy. To overcome limitations of small training data, we leverage an additional dataset of 2,029 patients who did not receive immunotherapy in a semi-supervised framework to learn intrinsic imaging phenotypes of the disease. We evaluated model performance in two independent cohorts of 81 patients treated with immunotherapy. RESULTS: The deep learning model achieved area under receiver operating characteristics curve (AUC) of 0.791 (95% CI 0.633-0.950) and 0.812 (95% CI 0.669-0.956) for predicting immunotherapy response in the internal and external validation cohorts. When combined with PD-L1 expression, the integrative model further improved the AUC by 4-7% in absolute terms. CONCLUSION: The deep learning model achieved promising performance for predicting immunotherapy response from routine clinical and image data. The proposed multi-modal approach is general and can incorporate other relevant information to further improve prediction of immunotherapy response.


Subject(s)
Deep Learning , Stomach Neoplasms , Humans , Immunotherapy , Phenotype , ROC Curve , Retrospective Studies
7.
Int J Surg ; 109(7): 2010-2024, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37300884

ABSTRACT

BACKGROUND: Peritoneal recurrence (PR) is the predominant pattern of relapse after curative-intent surgery in gastric cancer (GC) and indicates a dismal prognosis. Accurate prediction of PR is crucial for patient management and treatment. The authors aimed to develop a noninvasive imaging biomarker from computed tomography (CT) for PR evaluation, and investigate its associations with prognosis and chemotherapy benefit. METHODS: In this multicenter study including five independent cohorts of 2005 GC patients, the authors extracted 584 quantitative features from the intratumoral and peritumoral regions on contrast-enhanced CT images. The artificial intelligence algorithms were used to select significant PR-related features, and then integrated into a radiomic imaging signature. And improvements of diagnostic accuracy for PR by clinicians with the signature assistance were quantified. Using Shapley values, the authors determined the most relevant features and provided explanations to prediction. The authors further evaluated its predictive performance in prognosis and chemotherapy response. RESULTS: The developed radiomics signature had a consistently high accuracy in predicting PR in the training cohort (area under the curve: 0.732) and internal and Sun Yat-sen University Cancer Center validation cohorts (0.721 and 0.728). The radiomics signature was the most important feature in Shapley interpretation. The diagnostic accuracy of PR with the radiomics signature assistance was improved by 10.13-18.86% for clinicians ( P <0.001). Furthermore, it was also applicable in the survival prediction. In multivariable analysis, the radiomics signature remained an independent predictor for PR and prognosis ( P <0.001 for all). Importantly, patients with predicting high risk of PR from radiomics signature could gain survival benefit from adjuvant chemotherapy. By contrast, chemotherapy had no impact on survival for patients with a predicted low risk of PR. CONCLUSION: The noninvasive and explainable model developed from preoperative CT images could accurately predict PR and chemotherapy benefit in patients with GC, which will allow the optimization of individual decision-making.


Subject(s)
Peritoneal Neoplasms , Stomach Neoplasms , Humans , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/drug therapy , Stomach Neoplasms/surgery , Artificial Intelligence , Peritoneal Neoplasms/diagnostic imaging , Peritoneal Neoplasms/drug therapy , Retrospective Studies , Neoplasm Recurrence, Local/diagnostic imaging , Gastrectomy
8.
J Immunother Cancer ; 11(11)2023 11 21.
Article in English | MEDLINE | ID: mdl-38179695

ABSTRACT

BACKGROUND: Despite remarkable benefits have been provided by immune checkpoint inhibitors in gastric cancer (GC), predictions of treatment response and prognosis remain unsatisfactory, making identifying biomarkers desirable. The aim of this study was to develop and validate a CT imaging biomarker to predict the immunotherapy response in patients with GC and investigate the associated immune infiltration patterns. METHODS: This retrospective study included 294 GC patients who received anti-PD-1/PD-L1 immunotherapy from three independent medical centers between January 2017 and April 2022. A radiomics score (RS) was developed from the intratumoral and peritumoral features on pretreatment CT images to predict immunotherapy-related progression-free survival (irPFS). The performance of the RS was evaluated by the area under the time-dependent receiver operating characteristic curve (AUC). Multivariable Cox regression analysis was performed to construct predictive nomogram of irPFS. The C-index was used to determine the performance of the nomogram. Bulk RNA sequencing of tumors from 42 patients in The Cancer Genome Atlas was used to investigate the RS-associated immune infiltration patterns. RESULTS: Overall, 89 of 294 patients (median age, 57 years (IQR 48-66 years); 171 males) had an objective response to immunotherapy. The RS included 13 CT features that yielded AUCs of 12-month irPFS of 0.787, 0.810 and 0.785 in the training, internal validation, and external validation 1 cohorts, respectively, and an AUC of 24-month irPFS of 0.805 in the external validation 2 cohort. Patients with low RS had longer irPFS in each cohort (p<0.05). Multivariable Cox regression analyses showed RS is an independent prognostic factor of irPFS. The nomogram that integrated the RS and clinical characteristics showed improved performance in predicting irPFS, with C-index of 0.687-0.778 in the training and validation cohorts. The CT imaging biomarker was associated with M1 macrophage infiltration. CONCLUSION: The findings of this prognostic study suggest that the non-invasive CT imaging biomarker can effectively predict immunotherapy outcomes in patients with GC and is associated with innate immune signaling, which can serve as a potential tool for individual treatment decisions.


Subject(s)
Immunotherapy , Stomach Neoplasms , Humans , Male , Middle Aged , Biomarkers , Retrospective Studies , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/drug therapy , Tomography, X-Ray Computed , Female , Aged
9.
World J Gastroenterol ; 28(26): 3232-3242, 2022 Jul 14.
Article in English | MEDLINE | ID: mdl-36051348

ABSTRACT

BACKGROUND: Recently, hepatic arterial infusion chemotherapy (HAIC) plus lenvatinib has been frequently used to treat unresectable hepatocellular carcinoma (uHCC) in China. In the clinic, the hepatic arteries of some patients shrink significantly during this treatment, leading to improved short-term efficacy. AIM: To investigate the relationship between the shrinkage of hepatic arteries and the short-term effect of HAIC plus lenvatinib treatment. METHODS: Sixty-seven participants with uHCC were enrolled in this retrospective study. The patients received HAIC every 3 wk, followed by oral lenvatinib after the first HAIC course. Hepatic artery diameters were measured on CT before treatment and after 1 and 2 mo of treatment. Meanwhile, the changes in tumor capillaries were also examined on pathological specimens before and after 1 mo of treatment. The antitumor response after 1, 3, and 6 mo of treatment was assessed using the modified Response Evaluation Criteria in Solid Tumors (mRECIST). The relationship between the changes in vessel diameters and the short-term effect of the combination treatment was evaluated by receiver-operating characteristic and logistic regression analyses. RESULTS: The hepatic artery diameters were all significantly decreased after 1 and 2 mo of treatment (P < 0.001), but there was no difference in the vessel diameters between 1 and 2 mo (P > 0.05). The microvessel density in the tumor lesions decreased significantly after 1 mo of combination treatment (P < 0.001). According to mRECIST, 46, 41, and 24 patients had complete or partial responses after 1, 3, and 6 mo of treatment, respectively, whereas 21, 21, and 32 patients had a stable or progressive disease at these times, respectively. Shrinkage of the tumor-feeding artery was significantly associated with the tumor response after 1, 3, and 6 mo of treatment (P < 0.001, P = 0.004, and P = 0.023, respectively); however, changes in other hepatic arteries were not significantly associated with the tumor response. Furthermore, shrinkage of the tumor-feeding artery was an independent factor for treatment efficacy (P = 0.001, P = 0.001, and P = 0.002 and 1, 3, and 6 mo, respectively). CONCLUSION: The hepatic arteries shrank rapidly after treatment with HAIC plus lenvatinib, and shrinkage of the tumor-feeding artery diameter was closely related to improved short-term efficacy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Hepatic Artery/diagnostic imaging , Hepatic Artery/pathology , Humans , Infusions, Intra-Arterial , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Phenylurea Compounds , Quinolines , Retrospective Studies , Treatment Outcome
10.
Nat Commun ; 13(1): 5095, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36042205

ABSTRACT

The tumor immune microenvironment (TIME) is associated with tumor prognosis and immunotherapy response. Here we develop and validate a CT-based radiomics score (RS) using 2272 gastric cancer (GC) patients to investigate the relationship between the radiomics imaging biomarker and the neutrophil-to-lymphocyte ratio (NLR) in the TIME, including its correlation with prognosis and immunotherapy response in advanced GC. The RS achieves an AUC of 0.795-0.861 in predicting the NLR in the TIME. Notably, the radiomics imaging biomarker is indistinguishable from the IHC-derived NLR status in predicting DFS and OS in each cohort (HR range: 1.694-3.394, P < 0.001). We find the objective responses of a cohort of anti-PD-1 immunotherapy patients is significantly higher in the low-RS group (60.9% and 42.9%) than in the high-RS group (8.1% and 14.3%). The radiomics imaging biomarker is a noninvasive method to evaluate TIME, and may correlate with prognosis and anti PD-1 immunotherapy response in GC patients.


Subject(s)
Stomach Neoplasms , Biomarkers , Humans , Immunotherapy , Lymphocytes/pathology , Neutrophils/pathology , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy , Tumor Microenvironment
11.
Cell Death Dis ; 13(7): 658, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35902562

ABSTRACT

Chemoresistance remains the primary challenge of clinical treatment of gastric cancer (GC), making the biomarkers of chemoresistance crucial for treatment decision. Our previous study has reported that p21-actived kinase 6 (PAK6) is a prognostic factor for selecting which patients with GC are resistant to 5-fluorouracil/oxaliplatin chemotherapy. However, the mechanistic role of PAK6 in chemosensitivity remains unknown. The present study identified PAK6 as an important modulator of the DNA damage response (DDR) and chemosensitivity in GC. Analysis of specimens from patients revealed significant associations between the expression of PAK6 and poorer stages, deeper invasion, more lymph node metastases, higher recurrence rates, and resistance to oxaliplatin. Cells exhibited chemosensitivity to oxaliplatin after knockdown of PAK6, but showed more resistant to oxaliplatin when overexpressing PAK6. Functionally, PAK6 mediates cancer chemoresistance by enhancing homologous recombination (HR) to facilitate the DNA double-strand break repair. Mechanistically, PAK6 moves into nucleus to promote the activation of ATR, thereby further activating downstream repair protein CHK1 and recruiting RAD51 from cytoplasm to the DNA damaged site to repair the broken DNA in GC. Activation of ATR is the necessary step for PAK6 mediated HR repair to protect GC cells from oxaliplatin-induced apoptosis, and ATR inhibitor (AZD6738) could block the PAK6-mediated HR repair, thereby reversing the resistance to oxaliplatin and even promoting the sensitivity to oxaliplatin regardless of high expression of PAK6. In conclusion, these findings indicate a novel regulatory mechanism of PAK6 in modulating the DDR and chemoresistance in GC and provide a reversal suggestion in clinical decision.


Subject(s)
Stomach Neoplasms , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Fluorouracil/therapeutic use , Homologous Recombination , Humans , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
12.
Lancet Digit Health ; 4(5): e340-e350, 2022 05.
Article in English | MEDLINE | ID: mdl-35461691

ABSTRACT

BACKGROUND: Peritoneal recurrence is the predominant pattern of relapse after curative-intent surgery for gastric cancer and portends a dismal prognosis. Accurate individualised prediction of peritoneal recurrence is crucial to identify patients who might benefit from intensive treatment. We aimed to develop predictive models for peritoneal recurrence and prognosis in gastric cancer. METHODS: In this retrospective multi-institution study of 2320 patients, we developed a multitask deep learning model for the simultaneous prediction of peritoneal recurrence and disease-free survival using preoperative CT images. Patients in the training cohort (n=510) and the internal validation cohort (n=767) were recruited from Southern Medical University, Guangzhou, China. Patients in the external validation cohort (n=1043) were recruited from Sun Yat-sen University Cancer Center, Guangzhou, China. We evaluated the prognostic accuracy of the model as well as its association with chemotherapy response. Furthermore, we assessed whether the model could improve the ability of clinicians to predict peritoneal recurrence. FINDINGS: The deep learning model had a consistently high accuracy in predicting peritoneal recurrence in the training cohort (area under the receiver operating characteristic curve [AUC] 0·857; 95% CI 0·826-0·889), internal validation cohort (0·856; 0·829-0·882), and external validation cohort (0·843; 0·819-0·866). When informed by the artificial intelligence (AI) model, the sensitivity and inter-rater agreement of oncologists for predicting peritoneal recurrence was improved. The model was able to predict disease-free survival in the training cohort (C-index 0·654; 95% CI 0·616-0·691), internal validation cohort (0·668; 0·643-0·693), and external validation cohort (0·610; 0·583-0·636). In multivariable analysis, the model predicted peritoneal recurrence and disease-free survival independently of clinicopathological variables (p<0·0001 for all). For patients with a predicted high risk of peritoneal recurrence and low survival, adjuvant chemotherapy was associated with improved disease-free survival in both stage II disease (hazard ratio [HR] 0·543 [95% CI 0·362-0·815]; p=0·003) and stage III disease (0·531 [0·432-0·652]; p<0·0001). By contrast, chemotherapy had no impact on disease-free survival for patients with a predicted low risk of peritoneal recurrence and high survival. For the remaining patients, the benefit of chemotherapy depended on stage: only those with stage III disease derived benefit from chemotherapy (HR 0·637 [95% CI 0·484-0·838]; p=0·001). INTERPRETATION: The deep learning model could allow accurate prediction of peritoneal recurrence and survival in patients with gastric cancer. Prospective studies are required to test the clinical utility of this model in guiding personalised treatment in combination with clinicopathological criteria. FUNDING: None.


Subject(s)
Deep Learning , Peritoneal Neoplasms , Stomach Neoplasms , Artificial Intelligence , Disease-Free Survival , Humans , Neoplasm Recurrence, Local/diagnostic imaging , Predictive Value of Tests , Retrospective Studies , Stomach Neoplasms/diagnostic imaging , Tomography, X-Ray Computed
13.
Radiother Oncol ; 165: 179-190, 2021 12.
Article in English | MEDLINE | ID: mdl-34774652

ABSTRACT

BACKGROUND: Specific diagnosis and treatment of gastric cancer (GC) require accurate preoperative predictions of lymph node metastasis (LNM) at individual stations, such as estimating the extent of lymph node dissection. This study aimed to develop a radiomics signature based on preoperative computed tomography (CT) images, for predicting the LNM status at each individual station. METHODS: We enrolled 1506 GC patients retrospectively from two centers as training (531) and external (975) validation cohorts, and recruited 112 patients prospectively from a single center as prospective validation cohort. Radiomics features were extracted from preoperative CT images and integrated with clinical characteristics to construct nomograms for LNM prediction at individual lymph node stations. Performance of the nomograms was assessed through calibration, discrimination and clinical usefulness. RESULTS: In training, external and prospective validation cohorts, radiomics signature was significantly associated with LNM status. Moreover, radiomics signature was an independent predictor of LNM status in the multivariable logistic regression analysis. The radiomics nomograms revealed good prediction performances, with AUCs of 0.716-0.871 in the training cohort, 0.678-0.768 in the external validation cohort and 0.700-0.841 in the prospective validation cohort for 12 nodal stations. The nomograms demonstrated a significant agreement between the actual probability and predictive probability in calibration curves. Decision curve analysis showed that nomograms had better net benefit than clinicopathologic characteristics. CONCLUSION: Radiomics nomograms for individual lymph node stations presented good prediction accuracy, which could provide important information for individual diagnosis and treatment of gastric cancer.


Subject(s)
Stomach Neoplasms , Humans , Lymph Nodes/diagnostic imaging , Lymphatic Metastasis/diagnostic imaging , Nomograms , Retrospective Studies , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/surgery , Tomography, X-Ray Computed
14.
Lancet Digit Health ; 3(6): e371-e382, 2021 06.
Article in English | MEDLINE | ID: mdl-34045003

ABSTRACT

BACKGROUND: The tumour stroma microenvironment plays an important part in disease progression and its composition can influence treatment response and outcomes. Histological evaluation of tumour stroma is limited by access to tissue, spatial heterogeneity, and temporal evolution. We aimed to develop a radiological signature for non-invasive assessment of tumour stroma and treatment outcomes. METHODS: In this multicentre, retrospective study, we analysed CT images and outcome data of 2209 patients with resected gastric cancer from five independent cohorts recruited from two centres (Nanfang Hospital of Southern Medical University [Guangzhou, China] and Sun Yat-sen University Cancer Center [Guangzhou, China]). Patients with histologically confirmed gastric cancer, at least 15 lymph nodes harvested, preoperative abdominal CT available, and complete clinicopathological and follow-up data were eligible for inclusion. Tumour tissue was collected for patients in the training cohort (321 patients), internal validation cohort one (246 patients), and external validation cohort one (128 patients). Four stroma classes were defined according to the protein expression of α-smooth muscle actin and periostin assessed by immunohistochemistry. The primary objective was to predict the histologically based stroma classes by using preoperative CT images. We trained a deep convolutional neural network model using the training cohort and tested the model in the internal and external validation cohort one. We evaluated the model's association with prognosis in the training cohort, two internal, and two external validation cohorts and compared outcomes of patients who received or did not receive adjuvant chemotherapy. FINDINGS: The deep-learning model achieved a high diagnostic accuracy for assessing tumour stroma in both internal validation cohort one (area under the receiver operating characteristic curve [AUC] 0·96-0·98]) and external validation cohort one (AUC 0·89-0·94). The stromal imaging signature was significantly associated with disease-free survival and overall survival in all cohorts (p<0·0001). The predicted stroma classes remained an independent prognostic factor adjusting for clinicopathological variables including tumour size, stage, differentiation, and Lauren histology. In patients with stage II or III disease in predicted stroma classes one and two subgroups, patients who received adjuvant chemotherapy had improved survival compared with those who did not (in those with stage II disease hazard ratio [HR] 0·48 [95% CI 0·29-0·77], p=0·0021; and in those with stage III disease HR 0·70 [0·57-0·85], p=0·00042). However, in the other two subgroups adjuvant chemotherapy was not associated with survival and might even be detrimental in the predicted stroma class 4 subgroup (HR 1·48 [1·08-2·03], p=0·013). INTERPRETATION: The deep-learning model could allow for accurate and non-invasive evaluation of tumour stroma from CT images in gastric cancer. The radiographical model predicted chemotherapy outcomes and could be used in combination with clinicopathological criteria to refine prognosis and inform treatment decisions of patients with gastric cancer. FUNDING: None.


Subject(s)
Deep Learning , Stomach Neoplasms/diagnosis , Stomach/pathology , Tomography, X-Ray Computed/methods , Area Under Curve , Biomarkers, Tumor/metabolism , Chemotherapy, Adjuvant , China , Disease-Free Survival , Female , Humans , Male , Middle Aged , Neural Networks, Computer , Predictive Value of Tests , Prognosis , Progression-Free Survival , Proportional Hazards Models , ROC Curve , Radiography , Retrospective Studies , Stomach Neoplasms/classification , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/pathology
15.
JAMA Netw Open ; 4(1): e2032269, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33399858

ABSTRACT

Importance: Occult peritoneal metastasis frequently occurs in patients with advanced gastric cancer and is poorly diagnosed with currently available tools. Because the presence of peritoneal metastasis precludes the possibility of curative surgery, there is an unmet need for a noninvasive approach to reliably identify patients with occult peritoneal metastasis. Objective: To assess the use of a deep learning model for predicting occult peritoneal metastasis based on preoperative computed tomography images. Design, Setting, and Participants: In this multicenter, retrospective cohort study, a deep convolutional neural network, the Peritoneal Metastasis Network (PMetNet), was trained to predict occult peritoneal metastasis based on preoperative computed tomography images. Data from a cohort of 1225 patients with gastric cancer who underwent surgery at Sun Yat-sen University Cancer Center (Guangzhou, China) were used for training purposes. To externally validate the model, data were collected from 2 independent cohorts comprising a total of 753 patients with gastric cancer who underwent surgery at Nanfang Hospital (Guangzhou, China) or the Third Affiliated Hospital of Southern Medical University (Guangzhou, China). The status of peritoneal metastasis for all patients was confirmed by pathological examination of pleural specimens obtained during surgery. Detailed clinicopathological data were collected for each patient. Data analysis was performed between September 1, 2019, and January 31, 2020. Main Outcomes and Measures: The area under the receiver operating characteristic curve (AUC) and decision curve were analyzed to evaluate performance in predicting occult peritoneal metastasis. Results: A total of 1978 patients (mean [SD] age, 56.0 [12.2] years; 1350 [68.3%] male) were included in the study. The PMetNet model achieved an AUC of 0.946 (95% CI, 0.927-0.965), with a sensitivity of 75.4% and a specificity of 92.9% in external validation cohort 1. In external validation cohort 2, the AUC was 0.920 (95% CI, 0.848-0.992), with a sensitivity of 87.5% and a specificity of 98.2%. The discrimination performance of PMetNet was substantially higher than conventional clinicopathological factors (AUC range, 0.51-0.63). In multivariable logistic regression analysis, PMetNet was an independent predictor of occult peritoneal metastasis. Conclusions and Relevance: The findings of this cohort study suggest that the PMetNet model can serve as a reliable noninvasive tool for early identification of patients with clinically occult peritoneal metastasis, which will inform individualized preoperative treatment decision-making and may avoid unnecessary surgery and complications. These results warrant further validation in prospective studies.


Subject(s)
Deep Learning , Peritoneal Neoplasms/diagnostic imaging , Peritoneal Neoplasms/secondary , Stomach Neoplasms/pathology , Tomography, X-Ray Computed , China , Female , Humans , Male , Middle Aged , Peritoneal Neoplasms/surgery , Predictive Value of Tests , Retrospective Studies , Sensitivity and Specificity , Stomach Neoplasms/surgery
16.
Ann Surg ; 274(6): e1153-e1161, 2021 12 01.
Article in English | MEDLINE | ID: mdl-31913871

ABSTRACT

OBJECTIVE: We aimed to develop a deep learning-based signature to predict prognosis and benefit from adjuvant chemotherapy using preoperative computed tomography (CT) images. BACKGROUND: Current staging methods do not accurately predict the risk of disease relapse for patients with gastric cancer. METHODS: We proposed a novel deep neural network (S-net) to construct a CT signature for predicting disease-free survival (DFS) and overall survival in a training cohort of 457 patients, and independently tested it in an external validation cohort of 1158 patients. An integrated nomogram was constructed to demonstrate the added value of the imaging signature to established clinicopathologic factors for individualized survival prediction. Prediction performance was assessed with respect to discrimination, calibration, and clinical usefulness. RESULTS: The DeLIS was associated with DFS and overall survival in the overall validation cohort and among subgroups defined by clinicopathologic variables, and remained an independent prognostic factor in multivariable analysis (P< 0.001). Integrating the imaging signature and clinicopathologic factors improved prediction performance, with C-indices: 0.792-0.802 versus 0.719-0.724, and net reclassification improvement 10.1%-28.3%. Adjuvant chemotherapy was associated with improved DFS in stage II patients with high-DeLIS [hazard ratio = 0.362 (95% confidence interval 0.149-0.882)] and stage III patients with high- and intermediate-DeLIS [hazard ratio = 0.611 (0.442-0.843); 0.633 (0.433-0.925)]. On the other hand, adjuvant chemotherapy did not affect survival for patients with low-DeLIS, suggesting a predictive effect (Pinteraction = 0.048, 0.016 for DFS in stage II and III disease). CONCLUSIONS: The proposed imaging signature improved prognostic prediction and could help identify patients most likely to benefit from adjuvant chemotherapy in gastric cancer.


Subject(s)
Deep Learning , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/drug therapy , Tomography, X-Ray Computed , Aged , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Male , Middle Aged , Neoplasm Staging , Nomograms , Predictive Value of Tests , Prognosis , Retrospective Studies , Stomach Neoplasms/pathology
17.
Clin Transl Gastroenterol ; 11(10): e00253, 2020 10.
Article in English | MEDLINE | ID: mdl-33108125

ABSTRACT

INTRODUCTION: Treatments for young patients with gastric cancer (GC) remain poorly defined, and their effects on survival are uncertain. We aimed to investigate the receipt of chemotherapy by age category (18-49, 50-64, and 65-85 years) and explore whether age differences in chemotherapy matched survival gains in patients with GC. METHODS: Patients who were histologically diagnosed with GC were included from a Chinese multi-institutional database and the Surveillance, Epidemiology, and End Results database. There were 5,122 and 31,363 patients aged 18-85 years treated between 2000 and 2014, respectively. Overall survival and stage-specific likelihood of receiving chemotherapy were evaluated. RESULTS: Of the 5,122 and 31,363 patients in China and Surveillance, Epidemiology, and End Result data sets, 3,489 (68.1%) and 18,115 (57.8%) were men, respectively. Younger (18-49 years) and middle-aged (50-64 years) patients were more likely to receive chemotherapy compared with older patients (65-85 years) (64.9%, 56.7%, and 45.4% in the 3 groups from the China data set). Among patients treated with surgery alone, a significantly better prognosis was found in younger and middle-aged patients than their older counterparts; however, no significant differences were found in overall survival among age subgroups in patients who received both surgery and chemotherapy, especially in the China data set. The survival benefit from chemotherapy was superior among older patients (all P < 0.0001) compared with that among younger and middle-aged patients in stage II and III disease. DISCUSSION: Potential overuse of chemotherapy was found in younger and middle-aged patients with GC, but the addition of chemotherapy did not bring about matched survival improvement, especially in the China data set.


Subject(s)
Antineoplastic Agents/therapeutic use , Gastrectomy/statistics & numerical data , Stomach Neoplasms/therapy , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Chemotherapy, Adjuvant/statistics & numerical data , China/epidemiology , Datasets as Topic , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Prognosis , SEER Program/statistics & numerical data , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Treatment Outcome , United States/epidemiology , Young Adult
18.
Front Oncol ; 10: 1416, 2020.
Article in English | MEDLINE | ID: mdl-32974149

ABSTRACT

Objective: The aim of this study is to evaluate whether radiomics imaging signatures based on computed tomography (CT) could predict peritoneal metastasis (PM) in gastric cancer (GC) and to develop a nomogram for preoperative prediction of PM status. Methods: We collected CT images of pathological T4 gastric cancer in 955 consecutive patients of two cancer centers to analyze the radiomics features retrospectively and then developed and validated the prediction model built from 292 quantitative image features in the training cohort and two validation cohorts. Lasso regression model was applied for selecting feature and constructing radiomics signature. Predicting model was developed by multivariable logistic regression analysis. Radiomics nomogram was developed by the incorporation of radiomics signature and clinical T and N stage. Calibration, discrimination, and clinical usefulness were used to evaluate the performance of the nomogram. Results: In training and validation cohorts, PM status was associated with the radiomics signature significantly. It was found that the radiomics signature was an independent predictor for peritoneal metastasis in multivariable logistic analysis. For training and internal and external validation cohorts, the area under the receiver operating characteristic curves (AUCs) of radiomics signature for predicting PM were 0.751 (95%CI, 0.703-0.799), 0.802 (95%CI, 0.691-0.912), and 0.745 (95%CI, 0.683-0.806), respectively. Furthermore, for training and internal and external validation cohorts, the AUCs of radiomics nomogram for predicting PM were 0.792 (95%CI, 0.748-0.836), 0.870 (95%CI, 0.795-0.946), and 0.815 (95%CI, 0.763-0.867), respectively. Conclusions: CT-based radiomics signature could predict peritoneal metastasis, and the radiomics nomogram can make a meaningful contribution for predicting PM status in GC patient preoperatively.

19.
J Cancer ; 11(3): 678-685, 2020.
Article in English | MEDLINE | ID: mdl-31942191

ABSTRACT

Object: The risk of lymph node positivity (LN+) in gastric cancer (GC) impacts therapeutic recommendations. The aim of this study was to quantify the effect of younger age on LN+. Methods: Data from a Chinese multi-institutional database and the US SEER database on stage I to III resected GC were analyzed for the relationship between age and LN+ status. The association of age and LN+ status was examined with logistic regression separately for each T stage, adjusting for multiple covariates. Poisson regression was used to evaluate age and number of LN+. Results: 4,905 and 14,877 patients were identified in the China and SEER datasets respectively. 479 (9.8%) patients were under age 40 years, with 768 (15.7%) between age 40 and 49 years in China dataset, and 416 (2.8%) patients were under age 40 years, with 1176 (7.9%) between age 40 and 49 years in SEER dataset. Both datasets exhibited significantly proportional decreases of N3a and N3b LN+ with age increasing. Patients younger than age 40 years were more likely to show LN+ compared with the reference age 60 to 69 years. The youngest patients had the highest ORs of N1, N2, N3a, and N3b vs N0 LN+ within T4 stage of China dataset and T3 stage of SEER dataset, the values of ORs decreased with increasing age. Young age was a predictor of an increased number of LNs positive for each T stage. Conclusion: In the two large datasets, young age at diagnosis is associated with an increased risk of LN+.

20.
Front Oncol ; 9: 340, 2019.
Article in English | MEDLINE | ID: mdl-31106158

ABSTRACT

Background: To evaluate whether radiomic feature-based computed tomography (CT) imaging signatures allow prediction of lymph node (LN) metastasis in gastric cancer (GC) and to develop a preoperative nomogram for predicting LN status. Methods: We retrospectively analyzed radiomics features of CT images in 1,689 consecutive patients from three cancer centers. The prediction model was developed in the training cohort and validated in internal and external validation cohorts. Lasso regression model was utilized to select features and build radiomics signature. Multivariable logistic regression analysis was utilized to develop the model. We integrated the radiomics signature, clinical T and N stage, and other independent clinicopathologic variables, and this was presented as a radiomics nomogram. The performance of the nomogram was assessed with calibration, discrimination, and clinical usefulness. Results: The radiomics signature was significantly associated with pathological LN stage in training and validation cohorts. Multivariable logistic analysis found the radiomics signature was an independent predictor of LN metastasis. The nomogram showed good discrimination and calibration. Conclusions: The newly developed radiomic signature was a powerful predictor of LN metastasis and the radiomics nomogram could facilitate the preoperative individualized prediction of LN status.

SELECTION OF CITATIONS
SEARCH DETAIL
...