Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Nat Commun ; 15(1): 4369, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778014

ABSTRACT

Cervical cancer is a significant global health issue, its prevalence and prognosis highlighting the importance of early screening for effective prevention. This research aimed to create and validate an artificial intelligence cervical cancer screening (AICCS) system for grading cervical cytology. The AICCS system was trained and validated using various datasets, including retrospective, prospective, and randomized observational trial data, involving a total of 16,056 participants. It utilized two artificial intelligence (AI) models: one for detecting cells at the patch-level and another for classifying whole-slide image (WSIs). The AICCS consistently showed high accuracy in predicting cytology grades across different datasets. In the prospective assessment, it achieved an area under curve (AUC) of 0.947, a sensitivity of 0.946, a specificity of 0.890, and an accuracy of 0.892. Remarkably, the randomized observational trial revealed that the AICCS-assisted cytopathologists had a significantly higher AUC, specificity, and accuracy than cytopathologists alone, with a notable 13.3% enhancement in sensitivity. Thus, AICCS holds promise as an additional tool for accurate and efficient cervical cancer screening.


Subject(s)
Artificial Intelligence , Early Detection of Cancer , Uterine Cervical Neoplasms , Humans , Female , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/pathology , Early Detection of Cancer/methods , Adult , Middle Aged , Prospective Studies , Retrospective Studies , Sensitivity and Specificity , Cervix Uteri/pathology , Neoplasm Grading , Area Under Curve , Cytology
2.
Aging (Albany NY) ; 16(9): 7818-7844, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38700505

ABSTRACT

BACKGROUND: Stomach cancer is a leading cause of cancer-related deaths globally due to its high grade and poor response to treatment. Understanding the molecular network driving the rapid progression of stomach cancer is crucial for improving patient outcomes. METHODS: This study aimed to investigate the role of unfolded protein response (UPR) related genes in stomach cancer and their potential as prognostic biomarkers. RNA expression data and clinical follow-up information were obtained from the TCGA and GEO databases. An unsupervised clustering algorithm was used to identify UPR genomic subtypes in stomach cancer. Functional enrichment analysis, immune landscape analysis, and chemotherapy benefit prediction were conducted for each subtype. A prognostic model based on UPR-related genes was developed and validated using LASSO-Cox regression, and a multivariate nomogram was created. Key gene expression analyses in pan-cancer and in vitro experiments were performed to further investigate the role of the identified genes in cancer progression. RESULTS: A total of 375 stomach cancer patients were included in this study. Analysis of 113 UPR-related genes revealed their close functional correlation and significant enrichment in protein modification, transport, and RNA degradation pathways. Unsupervised clustering identified two molecular subtypes with significant differences in prognosis and gene expression profiles. Immune landscape analysis showed that UPR may influence the composition of the tumor immune microenvironment. Chemotherapy sensitivity analysis indicated that patients in the C2 molecular subtype were more responsive to chemotherapy compared to those in the C1 molecular subtype. A prognostic signature consisting of seven UPR-related genes was constructed and validated, and an independent prognostic nomogram was developed. The gene IGFBP1, which had the highest weight coefficient in the prognostic signature, was found to promote the malignant phenotype of stomach cancer cells, suggesting its potential as a therapeutic target. CONCLUSIONS: The study developed a UPR-related gene classifier and risk signature for predicting survival in stomach cancer, identifying IGFBP1 as a key factor promoting the disease's malignancy and a potential therapeutic target. IGFBP1's role in enhancing cancer cell adaptation to endoplasmic reticulum stress suggests its importance in stomach cancer prognosis and treatment.


Subject(s)
Biomarkers, Tumor , Stomach Neoplasms , Tumor Microenvironment , Unfolded Protein Response , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Unfolded Protein Response/genetics , Unfolded Protein Response/immunology , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Female , Male , Nomograms , Transcriptome , Gene Expression Profiling , Middle Aged
3.
Front Genet ; 15: 1332935, 2024.
Article in English | MEDLINE | ID: mdl-38756447

ABSTRACT

Background: In breast cancer oncogenesis, the precise role of cell apoptosis holds untapped potential for prognostic and therapeutic insights. Thus, it is important to develop a model predicated for breast cancer patients' prognosis and immunotherapy response based on apoptosis-related signature. Methods: Our approach involved leveraging a training dataset from The Cancer Genome Atlas (TCGA) to construct an apoptosis-related gene prognostic model. The model's validity was then tested across several cohorts, including METABRIC, Sun Yat-sen Memorial Hospital Sun Yat-sen University (SYSMH), and IMvigor210, to ensure its applicability and robustness across different patient demographics and treatment scenarios. Furthermore, we utilized Quantitative Polymerase Chain Reaction (qPCR) analysis to explore the expression patterns of these model genes in breast cancer cell lines compared to immortalized mammary epithelial cell lines, aiming to confirm their differential expression and underline their significance in the context of breast cancer. Results: Through the development and validation of our prognostic model based on seven apoptosis-related genes, we have demonstrated its substantial predictive power for the survival outcomes of breast cancer patients. The model effectively stratified patients into high and low-risk categories, with high-risk patients showing significantly poorer overall survival in the training cohort and across all validation cohorts. Importantly, qPCR analysis confirmed that the genes constituting our model indeed exhibit differential expression in breast cancer cell lines when contrasted with immortalized mammary epithelial cell lines. Conclusion: Our study establishes a groundbreaking prognostic model using apoptosis-related genes to enhance the precision of breast cancer prognosis and treatment, particularly in predicting immunotherapy response.

4.
Nanoscale Adv ; 6(8): 1974-1991, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38633037

ABSTRACT

Sonodynamic therapy (SDT) is an emerging approach for malignant tumor treatment, offering high precision, deep tissue penetration, and minimal side effects. The rapid advancements in nanotechnology, particularly in cancer treatment, have enhanced the efficacy and targeting specificity of SDT. Combining sonodynamic therapy with nanotechnology offers a promising direction for future cancer treatments. In this review, we first systematically discussed the anti-tumor mechanism of SDT and then summarized the common nanotechnology-related sonosensitizers and their recent applications. Subsequently, nanotechnology-related therapies derived using the SDT mechanism were elaborated. Finally, the role of nanomaterials in SDT combined therapy was also introduced.

5.
Heliyon ; 10(5): e27151, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38495207

ABSTRACT

The development of immune checkpoint inhibitors (ICIs) has significantly advanced cancer treatment. However, their efficacy is not consistent across all patients, underscoring the need for personalized approaches. In this study, we examined the relationship between activated CD4+ memory T cell expression and ICI responsiveness. A notable correlation was observed between increased activated CD4+ memory T cell expression and better patient survival in various cohorts. Additionally, the chemokine CXCL13 was identified as a potential prognostic biomarker, with higher expression levels associated with improved outcomes. Further analysis highlighted CXCL13's role in influencing the Tumor Microenvironment, emphasizing its relevance in tumor immunity. Using these findings, we developed a deep learning model by the Multi-Layer Aggregation Graph Neural Network method. This model exhibited promise in predicting ICI treatment efficacy, suggesting its potential application in clinical practice.

6.
MedComm (2020) ; 5(3): e471, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38434763

ABSTRACT

The exact function of M1 macrophages and CXCL9 in forecasting the effectiveness of immune checkpoint inhibitors (ICIs) is still not thoroughly investigated. We investigated the potential of M1 macrophage and C-X-C Motif Chemokine Ligand 9 (CXCL9) as predictive markers for ICI efficacy, employing a comprehensive approach integrating multicohort analysis and single-cell RNA sequencing. A significant correlation between high M1 macrophage and improved overall survival (OS) and objective response rate (ORR) was found. M1 macrophage expression was most pronounced in the immune-inflamed phenotype, aligning with increased expression of immune checkpoints. Furthermore, CXCL9 was identified as a key marker gene that positively correlated with M1 macrophage and response to ICIs, while also exhibiting associations with immune-related pathways and immune cell infiltration. Additionally, through exploring RNA epigenetic modifications, we identified Apolipoprotein B MRNA Editing Enzyme Catalytic Subunit 3G (APOBEC3G) as linked to ICI response, with high expression correlating with improved OS and immune-related pathways. Moreover, a novel model based on M1 macrophage, CXCL9, and APOBEC3G-related genes was developed using multi-level attention graph neural network, which showed promising predictive ability for ORR. This study illuminates the pivotal contributions of M1 macrophages and CXCL9 in shaping an immune-active microenvironment, correlating with enhanced ICI efficacy. The combination of M1 macrophage, CXCL9, and APOBEC3G provides a novel model for predicting clinical outcomes of ICI therapy, facilitating personalized immunotherapy.

7.
Int J Surg ; 110(5): 2604-2613, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38348891

ABSTRACT

OBJECTIVES: The authors aimed to assess the performance of a deep learning (DL) model, based on a combination of ultrasound (US) and mammography (MG) images, for predicting malignancy in breast lesions categorized as Breast Imaging Reporting and Data System (BI-RADS) US 4A in diagnostic patients with dense breasts. METHODS: A total of 992 patients were randomly allocated into the training cohort and the test cohort at a proportion of 4:1. Another, 218 patients were enrolled to form a prospective validation cohort. The DL model was developed by incorporating both US and MG images. The predictive performance of the combined DL model for malignancy was evaluated by sensitivity, specificity, and area under the receiver operating characteristic curve (AUC). The combined DL model was then compared to a clinical nomogram model and to the DL model trained using US image only and to that trained MG image only. RESULTS: The combined DL model showed satisfactory diagnostic performance for predicting malignancy in breast lesions, with an AUC of 0.940 (95% CI: 0.874-1.000) in the test cohort, and an AUC of 0.906 (95% CI: 0.817-0.995) in the validation cohort, which was significantly higher than the clinical nomogram model, and the DL model for US or MG alone ( P <0.05). CONCLUSIONS: The study developed an objective DL model combining both US and MG imaging features, which was proven to be more accurate for predicting malignancy in the BI-RADS US 4A breast lesions of patients with dense breasts. This model may then be used to more accurately guide clinicians' choices about whether performing biopsies in breast cancer diagnosis.


Subject(s)
Breast Density , Breast Neoplasms , Deep Learning , Mammography , Ultrasonography, Mammary , Humans , Female , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Middle Aged , Adult , Prospective Studies , Aged , Breast/diagnostic imaging , Breast/pathology , Sensitivity and Specificity , ROC Curve , Predictive Value of Tests
8.
Eur Radiol ; 34(3): 1774-1789, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37658888

ABSTRACT

OBJECTIVES: Accurate preoperative estimation of the risk of breast-conserving surgery (BCS) resection margin positivity would be beneficial to surgical planning. In this multicenter validation study, we developed an MRI-based radiomic model to predict the surgical margin status. METHODS: We retrospectively collected preoperative breast MRI of patients undergoing BCS from three hospitals (SYMH, n = 296; SYSUCC, n = 131; TSPH, n = 143). Radiomic-based model for risk prediction of the margin positivity was trained on the SYMH patients (7:3 ratio split for the training and testing cohorts), and externally validated in the SYSUCC and TSPH cohorts. The model was able to stratify patients into different subgroups with varied risk of margin positivity. Moreover, we used the immune-radiomic models and epithelial-mesenchymal transition (EMT) signature to infer the distribution patterns of immune cells and tumor cell EMT status under different marginal status. RESULTS: The AUCs of the radiomic-based model were 0.78 (0.66-0.90), 0.88 (0.79-0.96), and 0.76 (0.68-0.84) in the testing cohort and two external validation cohorts, respectively. The actual margin positivity rates ranged between 0-10% and 27.3-87.2% in low-risk and high-risk subgroups, respectively. Positive surgical margin was associated with higher levels of EMT and B cell infiltration in the tumor area, as well as the enrichment of B cells, immature dendritic cells, and neutrophil infiltration in the peritumoral area. CONCLUSIONS: This MRI-based predictive model can be used as a reliable tool to predict the risk of margin positivity of BCS. Tumor immune-microenvironment alteration was associated with surgical margin status. CLINICAL RELEVANCE STATEMENT: This study can assist the pre-operative planning of BCS. Further research on the tumor immune microenvironment of different resection margin states is expected to develop new margin evaluation indicators and decipher the internal mechanism. KEY POINTS: • The MRI-based radiomic prediction model (CSS model) incorporating features extracted from multiple sequences and segments could estimate the margin positivity risk of breast-conserving surgery. • The radiomic score of the CSS model allows risk stratification of patients undergoing breast-conserving surgery, which could assist in surgical planning. • With the help of MRI-based radiomics to estimate the components of the immune microenvironment, for the first time, it is found that the margin status of breast-conserving surgery is associated with the infiltration of immune cells in the microenvironment and the EMT status of breast tumor cells.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/surgery , Breast Neoplasms/pathology , Mastectomy, Segmental , Margins of Excision , Retrospective Studies , Radiomics , Magnetic Resonance Imaging , Tumor Microenvironment
9.
Breast Cancer Res ; 25(1): 132, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37915093

ABSTRACT

BACKGROUND: Several studies have indicated that magnetic resonance imaging radiomics can predict survival in patients with breast cancer, but the potential biological underpinning remains indistinct. Herein, we aim to develop an interpretable deep-learning-based network for classifying recurrence risk and revealing the potential biological mechanisms. METHODS: In this multicenter study, 1113 nonmetastatic invasive breast cancer patients were included, and were divided into the training cohort (n = 698), the validation cohort (n = 171), and the testing cohort (n = 244). The Radiomic DeepSurv Net (RDeepNet) model was constructed using the Cox proportional hazards deep neural network DeepSurv for predicting individual recurrence risk. RNA-sequencing was performed to explore the association between radiomics and tumor microenvironment. Correlation and variance analyses were conducted to examine changes of radiomics among patients with different therapeutic responses and after neoadjuvant chemotherapy. The association and quantitative relation of radiomics and epigenetic molecular characteristics were further analyzed to reveal the mechanisms of radiomics. RESULTS: The RDeepNet model showed a significant association with recurrence-free survival (RFS) (HR 0.03, 95% CI 0.02-0.06, P < 0.001) and achieved AUCs of 0.98, 0.94, and 0.92 for 1-, 2-, and 3-year RFS, respectively. In the validation and testing cohorts, the RDeepNet model could also clarify patients into high- and low-risk groups, and demonstrated AUCs of 0.91 and 0.94 for 3-year RFS, respectively. Radiomic features displayed differential expression between the two risk groups. Furthermore, the generalizability of RDeepNet model was confirmed across different molecular subtypes and patient populations with different therapy regimens (All P < 0.001). The study also identified variations in radiomic features among patients with diverse therapeutic responses and after neoadjuvant chemotherapy. Importantly, a significant correlation between radiomics and long non-coding RNAs (lncRNAs) was discovered. A key lncRNA was found to be noninvasively quantified by a deep learning-based radiomics prediction model with AUCs of 0.79 in the training cohort and 0.77 in the testing cohort. CONCLUSIONS: This study demonstrates that machine learning radiomics of MRI can effectively predict RFS after surgery in patients with breast cancer, and highlights the feasibility of non-invasive quantification of lncRNAs using radiomics, which indicates the potential of radiomics in guiding treatment decisions.


Subject(s)
Breast Neoplasms , RNA, Long Noncoding , Humans , Female , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/genetics , Breast Neoplasms/surgery , RNA, Long Noncoding/genetics , Machine Learning , Magnetic Resonance Imaging , Receptor Protein-Tyrosine Kinases , Cohort Studies , Retrospective Studies , Tumor Microenvironment
10.
Acta Pharm Sin B ; 13(8): 3489-3502, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37655325

ABSTRACT

Long non-coding RNAs (lncRNAs) play an important role in cancer metastasis. Exploring metastasis-associated lncRNAs and developing effective strategy for targeted regulation of lncRNA function in vivo are of utmost importance for the treatment of metastatic cancer, which however remains a big challenge. Herein, we identified a new functional lncRNA (denoted lncBCMA), which could stabilize the expression of eukaryotic translation elongation factor 1A1 (eEF1A1) via antagonizing its ubiquitination to promote triple-negative breast cancer (TNBC) growth and metastasis. Based on this regulatory mechanism, an endosomal pH-responsive nanoparticle (NP) platform was engineered for systemic lncBCMA siRNA (siBCMA) delivery. This NPs-mediated siBCMA delivery could effectively silence lncBCMA expression and promote eEF1A1 ubiquitination, thereby leading to a significant inhibition of TNBC tumor growth and metastasis. These findings show that lncBCMA could be used as a potential biomarker to predict the prognosis of TNBC patients and NPs-mediated lncBCMA silencing could be an effective strategy for metastatic TNBC treatment.

11.
Transl Oncol ; 37: 101738, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37597296

ABSTRACT

BACKGROUND: This study aimed to comprehensively explore the clinical significance of PIK3CA mutation in human epidermal growth factor receptor 2 (HER2)-positive breast cancer treated with anti-HER2 therapy. METHODS: We systematically searched PubMed, Embase, and the Cochrane databases for eligible studies assessing the association between PIK3CA mutation and outcomes in patients with HER2-positive breast cancer receiving anti-HER2 therapy. The main outcomes included: (1) pathological complete response (pCR) or disease-free survival (DFS) for the neoadjuvant setting; (2) DFS or invasive DFS for the adjuvant setting; (3) objective response rate (ORR), progression-free survival (PFS), time-to-progression (TTP), or overall survival (OS) for the metastatic setting. The mutational landscape of HER2-positive breast cancer according to PIK3CA mutation status was examined based on TCGA breast cancer dataset. RESULTS: Totally, 43 eligible studies, covering 11,099 patients with available data on PIK3CA mutation status, were identified. In the neoadjuvant setting, PIK3CA mutation was significantly associated with a lower pCR rate (OR=0.23, 95% CI 0.19-0.27, p<0.001). This association remained significant irrespective of the type of anti-HER2 therapy (single-agent or dual-agent) and hormone receptor status. There were no significant differences in DFS between PIK3CA mutated and wild-type patients in either the neoadjuvant or adjuvant settings. In the metastatic setting, PIK3CA mutation predicted worse ORR (OR=0.26, 95%CI 0.17-0.40, p<0.001), PFS (HR=1.28, 95%CI 1.03-1.59, p = 0.024) and TTP (HR=2.27, 95%CI 1.54-3.34, p<0.001). However, no significant association was observed between PIK3CA mutation status and OS. Distinct mutational landscapes were observed in HER2-positive breast cancer between individuals with PIK3CA mutations and those with wild-type PIK3CA. CONCLUSIONS: PIK3CA mutation was significantly associated with a lower pCR rate in HER2-positive breast cancer treated with neoadjuvant anti-HER2 therapy. In the metastatic setting, PIK3CA mutation was predictive of worse ORR, PFS and TTP. These results suggest the potential for developing PI3K inhibitors as a therapeutic option for these patients.

12.
Breast ; 71: 1-12, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37429049

ABSTRACT

INTRODUCTION: The relationships between body mass index (BMI) and survival rates are complex, and have not been thoroughly investigated in breast cancer patients who received adjuvant chemotherapy. METHODS: We collected data on 2394 patients from two randomized, phase III clinical trials that investigated adjuvant chemotherapy in breast cancer identified in Project Data Sphere. The objective was to examine the effect of baseline BMI, BMI after adjuvant chemotherapy, and BMI change from baseline to post-adjuvant chemotherapy on disease-free survival (DFS) and overall survival (OS). Restricted cubic splines were used to examine potential non-linear associations between continuous BMI value and survival. Stratified analyses involved chemotherapy regimens. RESULTS: Severe obesity (BMI≥40.0 kg/m2) at baseline was independently associated with worse DFS (hazard ration [HR] = 1.48, 95% confidence interval [CI] 1.02-2.16, P = 0.04) and OS (HR = 1.79, 95%CI 1.17-2.74, P = 0.007) compared with underweight/normal weight (BMI≤24.9 kg/m2). A BMI loss >10% was also an independent prognostic factor for adverse OS (HR = 2.14, 95%CI 1.17-3.93, P = 0.014). Stratified analyses revealed that severe obesity adversely affected DFS (HR = 2.38, 95%CI 1.26-4.34, P = 0.007) and OS (HR = 2.90, 95%CI 1.46-5.76, P = 0.002) in the docetaxel-based group, but not in the non-docetaxel-based group. Restricted cubic splines revealed a "J-shaped" association of baseline BMI with risk of recurrence or all-cause death, and this relationship was more pronounced in the docetaxel-based group. CONCLUSIONS: In early breast cancer patients treated with adjuvant chemotherapy, baseline severe obesity was significantly linked to worse DFS and OS, and a BMI loss over 10% from baseline to post-adjuvant chemotherapy also negatively affected OS. Moreover, the prognostic role of BMI might differ between docetaxel-based and non-docetaxel-based groups.


Subject(s)
Breast Neoplasms , Obesity, Morbid , Humans , Female , Body Mass Index , Obesity, Morbid/complications , Obesity, Morbid/drug therapy , Docetaxel/therapeutic use , Prognosis , Disease-Free Survival , Obesity/complications , Chemotherapy, Adjuvant , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
13.
J Immunother Cancer ; 11(5)2023 05.
Article in English | MEDLINE | ID: mdl-37217246

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs)-based therapy, is regarded as one of the major breakthroughs in cancer treatment. However, it is challenging to accurately identify patients who may benefit from ICIs. Current biomarkers for predicting the efficacy of ICIs require pathological slides, and their accuracy is limited. Here we aim to develop a radiomics model that could accurately predict response of ICIs for patients with advanced breast cancer (ABC). METHODS: Pretreatment contrast-enhanced CT (CECT) image and clinicopathological features of 240 patients with ABC who underwent ICIs-based treatment in three academic hospitals from February 2018 to January 2022 were assigned into a training cohort and an independent validation cohort. For radiomic features extraction, CECT images of patients 1 month prior to ICIs-based therapies were first delineated with regions of interest. Data dimension reduction, feature selection and radiomics model construction were carried out with multilayer perceptron. Combined the radiomics signatures with independent clinicopathological characteristics, the model was integrated by multivariable logistic regression analysis. RESULTS: Among the 240 patients, 171 from Sun Yat-sen Memorial Hospital and Sun Yat-sen University Cancer Center were evaluated as a training cohort, while other 69 from Sun Yat-sen University Cancer Center and the First Affiliated Hospital of Sun Yat-sen University were the validation cohort. The area under the curve (AUC) of radiomics model was 0.994 (95% CI: 0.988 to 1.000) in the training and 0.920 (95% CI: 0.824 to 1.000) in the validation set, respectively, which were significantly better than the performance of clinical model (0.672 for training and 0.634 for validation set). The integrated clinical-radiomics model showed increased but not statistical different predictive ability in both the training (AUC=0.997, 95% CI: 0.993 to 1.000) and validation set (AUC=0.961, 95% CI: 0.885 to 1.000) compared with the radiomics model. Furthermore, the radiomics model could divide patients under ICIs-therapies into high-risk and low-risk group with significantly different progression-free survival both in training (HR=2.705, 95% CI: 1.888 to 3.876, p<0.001) and validation set (HR=2.625, 95% CI: 1.506 to 4.574, p=0.001), respectively. Subgroup analyses showed that the radiomics model was not influenced by programmed death-ligand 1 status, tumor metastatic burden or molecular subtype. CONCLUSIONS: This radiomics model provided an innovative and accurate way that could stratify patients with ABC who may benefit more from ICIs-based therapies.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Biomarkers , Machine Learning
14.
Heliyon ; 9(3): e14450, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36950600

ABSTRACT

Although immunotherapy has revolutionized cancer management, most patients do not derive benefits from it. Aiming to explore an appropriate strategy for immunotherapy efficacy prediction, we collected 6251 patients' transcriptome data from multicohort population and analyzed the data using a machine learning algorithm. In this study, we found that patients from three immune gene clusters had different overall survival when treated with immunotherapy (P < 0.001), and that these clusters had differential states of hypoxia scores and metabolism functions. The immune gene score showed good immunotherapy efficacy prediction (AUC was 0.737 at 20 months), which was well validated. The immune gene score, tumor mutation burden, and long non-coding RNA score were further combined to build a tumor immune microenvironment signature, which correlated more strongly with overall survival (AUC, 0.814 at 20 months) than when using a single variable. Thus, we recommend using the characterization of the tumor immune microenvironment associated with immunotherapy efficacy via a multi-omics analysis of cancer.

15.
J Pers Med ; 13(3)2023 Mar 06.
Article in English | MEDLINE | ID: mdl-36983658

ABSTRACT

Immune checkpoint inhibitors (ICIs) represent a new hot spot in tumor therapy. Programmed cell death has an important role in the prognosis. We explore a programmed cell death gene prognostic model associated with survival and immunotherapy prediction via computational algorithms. Patient details were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus databases. We used LASSO algorithm and multiple-cox regression to establish a programmed cell death-associated gene prognostic model. Further, we explored whether this model could evaluate the sensitivity of patients to anti-PD-1/PD-L1. In total, 1342 patients were included. We constructed a programmed cell death model in TCGA cohorts, and the overall survival (OS) was significantly different between the high- and low-risk score groups (HR 2.70; 95% CI 1.94-3.75; p < 0.0001; 3-year OS AUC 0.71). Specifically, this model was associated with immunotherapy progression-free survival benefit in the validation cohort (HR 2.42; 95% CI 1.59-3.68; p = 0.015; 12-month AUC 0.87). We suggest that the programmed cell death model could provide guidance for immunotherapy in LUAD patients.

16.
Arch Gynecol Obstet ; 307(3): 891-901, 2023 03.
Article in English | MEDLINE | ID: mdl-35708782

ABSTRACT

PURPOSE: To evaluate the effects of adjuvant chemotherapy (CT) and radiotherapy (RT) on the survival of uterine carcinosarcoma (UCS) patients. METHODS: We analyzed 3207 patients with uterine carcinosarcoma without distant metastasis after surgery from 2004 to 2015 by utilizing data from the Surveillance, Epidemiology, and End Results database. Generally, cancer-specific survival (CSS) and overall survival (OS) outcomes were analyzed by Kaplan-Meier and Cox proportional hazards regression models. Further subgroup survival analysis was performed for those receiving RT and chemoradiotherapy (CRT). RESULTS: In general, both univariate and multivariate analyses showed that age, race, marital status, stage, lymph node metastasis, lymphadenectomy (LND), RT, and chemotherapy (CT) were associated with improved CSS and OS (P < 0.05). Further subgroup analysis showed that CRT exhibited a survival advantage over RT or CT alone in different groups. Various RT modalities, including brachytherapy (BT), external radiotherapy (EBRT), and EBRT + BT, were correlated with improved survival for patients aged 60-69 years with stage III-IV disease and lymph node metastasis. Patients with stage I-II disease aged > 70 years seemed to gain survival benefits from brachytherapy (BT) alone. BT with or without external radiotherapy was associated with improved survival for those who did not undergo lymphadenectomy. CONCLUSION: For UCS without distant metastasis after surgery, CRT should be considered. Regarding RT, BT alone is efficient in improving survival, especially for patients with stage I-II disease aged > 70 years old. EBRT alone does not show results in survival improvement for patients who did not undergo LND and those with lymph node metastasis. However, considering the limitation of SEER database, further studies with more large sample size and strict study design are needed to confirm it.


Subject(s)
Carcinosarcoma , Uterine Neoplasms , Female , Humans , Aged , Lymphatic Metastasis , Radiotherapy, Adjuvant/methods , Neoplasm Staging , Uterine Neoplasms/pathology , Chemotherapy, Adjuvant , Carcinosarcoma/pathology , Retrospective Studies
17.
Front Oncol ; 12: 895177, 2022.
Article in English | MEDLINE | ID: mdl-36505880

ABSTRACT

Objective: The aim of the study is to develop and validate a deep learning model to predict the platinum sensitivity of patients with epithelial ovarian cancer (EOC) based on contrast-enhanced magnetic resonance imaging (MRI). Methods: In this retrospective study, 93 patients with EOC who received platinum-based chemotherapy (≥4 cycles) and debulking surgery at the Sun Yat-sen Memorial Hospital from January 2011 to January 2020 were enrolled and randomly assigned to the training and validation cohorts (2:1). Two different models were built based on either the primary tumor or whole volume of the abdomen as the volume of interest (VOI) within the same cohorts, and then a pre-trained convolutional neural network Med3D (Resnet 10 version) was transferred to automatically extract 1,024 features from two MRI sequences (CE-T1WI and T2WI) of each patient to predict platinum sensitivity. The performance of the two models was compared. Results: A total of 93 women (mean age, 50.5 years ± 10.5 [standard deviation]) were evaluated (62 in the training cohort and 31 in the validation cohort). The AUCs of the whole abdomen model were 0.97 and 0.98 for the training and validation cohorts, respectively, which was better than the primary tumor model (AUCs of 0.88 and 0.81 in the training and validation cohorts, respectively). In k-fold cross-validation and stratified analysis, the whole abdomen model maintained a stable performance, and the decision function value generated by the model was a prognostic indicator that successfully discriminates high- and low-risk recurrence patients. Conclusion: The non-manually segmented whole-abdomen deep learning model based on MRI exhibited satisfactory predictive performance for platinum sensitivity and may assist gynecologists in making optimal treatment decisions.

18.
Healthcare (Basel) ; 10(11)2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36360490

ABSTRACT

Background: Sepsis commonly causes acute respiratory distress syndrome (ARDS), and ARDS contributes to poor prognosis in sepsis patients. Early prediction of ARDS for sepsis patients remains a clinical challenge. This study aims to develop and validate chest computed tomography (CT) radiomic-based signatures for early prediction of ARDS and assessment of individual severity in sepsis patients. Methods: In this ambispective observational cohort study, a deep learning model, a sepsis-induced acute respiratory distress syndrome (SI-ARDS) prediction neural network, will be developed to extract radiomics features of chest CT from sepsis patients. The datasets will be collected from these retrospective and prospective cohorts, including 400 patients diagnosed with sepsis-3 definition during a period from 1 May 2015 to 30 May 2022. 160 patients of the retrospective cohort will be selected as a discovering group to reconstruct the model and 40 patients of the retrospective cohort will be selected as a testing group for internal validation. Additionally, 200 patients of the prospective cohort from two hospitals will be selected as a validating group for external validation. Data pertaining to chest CT, clinical information, immune-associated inflammatory indicators and follow-up will be collected. The primary outcome is to develop and validate the model, predicting in-hospital incidence of SI-ARDS. Finally, model performance will be evaluated using the area under the curve (AUC) of receiver operating characteristic (ROC), sensitivity and specificity, using internal and external validations. Discussion: Present studies reveal that early identification and classification of the SI-ARDS is essential to improve prognosis and disease management. Chest CT has been sought as a useful diagnostic tool to identify ARDS. However, when characteristic imaging findings were clearly presented, delays in diagnosis and treatment were impossible to avoid. In this ambispective cohort study, we hope to develop a novel model incorporating radiomic signatures and clinical signatures to provide an easy-to-use and individualized prediction of SI-ARDS occurrence and severe degree in patients at early stage.

20.
Ann Surg Oncol ; 29(12): 7685-7693, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35773561

ABSTRACT

PURPOSE: This study aimed to identify patients with pathological complete response (pCR) and make better clinical decisions by constructing a preoperative predictive model based on tumoral and peritumoral volumes of multiparametric magnetic resonance imaging (MRI) obtained before neoadjuvant chemotherapy (NAC). METHODS: This study investigated MRI before NAC in 448 patients with nonmetastatic invasive ductal breast cancer (Sun Yat-sen Memorial Hospital, Sun Yat-sen University, n = 362, training cohort; and Sun Yat-sen University Cancer Center, n = 86, validation cohort). The tumoral and peritumoral volumes of interest (VOIs) were segmented and MRI features were extracted. The radiomic features were filtered via a random forest algorithm, and a supporting vector machine was used for modeling. The receiver operator characteristic curve and area under the curve (AUC) were calculated to assess the performance of the radiomics-based classifiers. RESULTS: For each MRI sequence, a total of 863 radiomic features were extracted and the top 30 features were selected for model construction. The radiomic classifiers of tumoral VOI and peritumoral VOI were both promising for predicting pCR, with AUCs of 0.96 and 0.97 in the training cohort and 0.89 and 0.78 in the validation cohort, respectively. The tumoral + peritumoral VOI radiomic model could further improve the predictive accuracy, with AUCs of 0.98 and 0.92 in the training and validation cohorts. CONCLUSIONS: The tumoral and peritumoral multiparametric MRI radiomics model can promisingly predict pCR in breast cancer using MRI images before surgery. Our results highlighted the potential value of the tumoral and peritumoral radiomic model in cancer management.


Subject(s)
Breast Neoplasms , Multiparametric Magnetic Resonance Imaging , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Female , Humans , Magnetic Resonance Imaging/methods , Neoadjuvant Therapy/methods , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...