Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Comput Biol Med ; 176: 108568, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744009

ABSTRACT

Discovery of the cancer type specific-driver genes is important for understanding the molecular mechanisms of each cancer type and for providing proper treatment. Recently, graph deep learning methods became widely used in finding cancer-driver genes. However, previous methods had limited performance in individual cancer types due to a small number of cancer-driver genes used in training and biases toward the cancer-driver genes used in training the models. Here, we introduce a novel pipeline, CancerGATE that predicts the cancer-driver genes using graph attention autoencoder (GATE) to learn in a self-supervised manner and can be applied to each of the cancer types. CancerGATE utilizes biological network topology and multi-omics data from 15 types of cancer of 20,079 samples from the cancer genome atlas (TCGA). Attention coefficients calculated in the model are used to prioritize cancer-driver genes by comparing coefficients of cancer and normal contexts. CancerGATE shows a higher AUPRC with a difference ranging from 1.5 % to 36.5 % compared to the previous graph deep learning models in each cancer type. We also show that CancerGATE is free from the bias toward cancer-driver genes used in training, revealing mechanisms of the cancer-driver genes in specific cancer types. Finally, we propose novel cancer-driver gene candidates that could be therapeutic targets for specific cancer types.


Subject(s)
Deep Learning , Neoplasms , Humans , Neoplasms/genetics , Computational Biology/methods , Software
2.
Gynecol Oncol ; 185: 194-201, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38452634

ABSTRACT

OBJECTIVE: Endometrial cancer (EndoCA) is the most common gynecologic cancer and incidence and mortality rate continue to increase. Despite well-characterized knowledge of EndoCA-defining mutations, no effective diagnostic or screening tests exist. To lay the foundation for testing development, our study focused on defining the prevalence of somatic mutations present in non-cancerous uterine tissue. METHODS: We obtained ≥8 uterine samplings, including separate endometrial and myometrial layers, from each of 22 women undergoing hysterectomy for non-cancer conditions. We ultra-deep sequenced (>2000× coverage) samples using a 125 cancer-relevant gene panel. RESULTS: All women harbored complex mutation patterns. In total, 308 somatic mutations were identified with mutant allele frequencies ranging up to 96.0%. These encompassed 56 unique mutations from 24 genes. The majority of samples possessed predicted functional cancer mutations but curiously no growth advantage over non-functional mutations was detected. Functional mutations were enriched with increasing patient age (p = 0.045) and BMI (p = 0.0007) and in endometrial versus myometrial layers (68% vs 39%, p = 0.0002). Finally, while the somatic mutation landscape shared similar mutation prevalence in key TCGA-defined EndoCA genes, notably PIK3CA, significant differences were identified, including NOTCH1 (77% vs 10%), PTEN (9% vs 61%), TP53 (0% vs 37%) and CTNNB1 (0% vs 26%). CONCLUSIONS: An important caveat for future liquid biopsy/DNA-based cancer diagnostics is the repertoire of shared and distinct mutation profiles between histologically unremarkable and EndoCA tissues. The lack of selection pressure between functional and non-functional mutations in histologically unremarkable uterine tissue may offer a glimpse into an unrecognized EndoCA protective mechanism.


Subject(s)
Endometrium , Mutation , Humans , Female , Middle Aged , Endometrium/pathology , Endometrium/metabolism , Aged , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Adult , High-Throughput Nucleotide Sequencing
3.
BMC Bioinformatics ; 25(1): 34, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38254011

ABSTRACT

BACKGROUND: Driver genes play a vital role in the development of cancer. Identifying driver genes is critical for diagnosing and understanding cancer. However, challenges remain in identifying personalized driver genes due to tumor heterogeneity of cancer. Although many computational methods have been developed to solve this problem, few efforts have been undertaken to explore gene-patient associations to identify personalized driver genes. RESULTS: Here we propose a method called LPDriver to identify personalized cancer driver genes by employing linear neighborhood propagation model on individual genetic data. LPDriver builds personalized gene network based on the genetic data of individual patients, extracts the gene-patient associations from the bipartite graph of the personalized gene network and utilizes a linear neighborhood propagation model to mine gene-patient associations to detect personalized driver genes. The experimental results demonstrate that as compared to the existing methods, our method shows competitive performance and can predict cancer driver genes in a more accurate way. Furthermore, these results also show that besides revealing novel driver genes that have been reported to be related with cancer, LPDriver is also able to identify personalized cancer driver genes for individual patients by their network characteristics even if the mutation data of genes are hidden. CONCLUSIONS: LPDriver can provide an effective approach to predict personalized cancer driver genes, which could promote the diagnosis and treatment of cancer. The source code and data are freely available at https://github.com/hyr0771/LPDriver .


Subject(s)
Neoplasms , Oncogenes , Humans , Mutation , Gene Regulatory Networks , Linear Models , Patients , Neoplasms/genetics
4.
Math Biosci Eng ; 20(12): 21643-21669, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38124614

ABSTRACT

Cancer driver genes (CDGs) are crucial in cancer prevention, diagnosis and treatment. This study employed computational methods for identifying CDGs, categorizing them into four groups. The major frameworks for each of these four categories were summarized. Additionally, we systematically gathered data from public databases and biological networks, and we elaborated on computational methods for identifying CDGs using the aforementioned databases. Further, we summarized the algorithms, mainly involving statistics and machine learning, used for identifying CDGs. Notably, the performances of nine typical identification methods for eight types of cancer were compared to analyze the applicability areas of these methods. Finally, we discussed the challenges and prospects associated with methods for identifying CDGs. The present study revealed that the network-based algorithms and machine learning-based methods demonstrated superior performance.


Subject(s)
Neoplasms , Humans , Neoplasms/diagnosis , Neoplasms/genetics , Algorithms , Computational Biology/methods
5.
Pharmaceuticals (Basel) ; 16(10)2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37895906

ABSTRACT

During tumorigenesis, urokinase (uPA) and uPA receptor (uPAR) play essential roles in mediating pathological progression in many cancers. To understand the crosstalk between the uPA/uPAR signaling and cancer, as well as to decipher their cellular pathways, we proposed to use cancer driver genes to map out the uPAR signaling. In the study, an integrated pharmaceutical bioinformatics approach that combined modulator identification, driver gene ontology networking, protein targets prediction and networking, pathway analysis and uPAR modulator screening platform construction was employed to uncover druggable targets in uPAR signaling for developing a novel anti-cancer modality. Through these works, we found that uPAR signaling interacted with 10 of 21 KEGG cancer pathways, indicating the important role of uPAR in mediating intracellular cancerous signaling. Furthermore, we verified that receptor tyrosine kinases (RTKs) and ribosomal S6 kinases (RSKs) could serve as signal hubs to relay uPAR-mediated cellular functions on cancer hallmarks such as angiogenesis, proliferation, migration and metastasis. Moreover, we established an in silico virtual screening platform and a uPAR-driver gene pair rule for identifying potential uPAR modulators to combat cancer. Altogether, our results not only elucidated the complex networking between uPAR modulation and cancer but also provided a paved way for developing new chemical entities and/or re-positioning clinically used drugs against cancer.

6.
Brief Bioinform ; 24(5)2023 09 20.
Article in English | MEDLINE | ID: mdl-37738403

ABSTRACT

Identifying personalized cancer driver genes and further revealing their oncogenic mechanisms is critical for understanding the mechanisms of cell transformation and aiding clinical diagnosis. Almost all existing methods primarily focus on identifying driver genes at the cohort or individual level but fail to further uncover their underlying oncogenic mechanisms. To fill this gap, we present an interpretable framework, PhenoDriver, to identify personalized cancer driver genes, elucidate their roles in cancer development and uncover the association between driver genes and clinical phenotypic alterations. By analyzing 988 breast cancer patients, we demonstrate the outstanding performance of PhenoDriver in identifying breast cancer driver genes at the cohort level compared to other state-of-the-art methods. Otherwise, our PhenoDriver can also effectively identify driver genes with both recurrent and rare mutations in individual patients. We further explore and reveal the oncogenic mechanisms of some known and unknown breast cancer driver genes (e.g. TP53, MAP3K1, HTT, etc.) identified by PhenoDriver, and construct their subnetworks for regulating clinical abnormal phenotypes. Notably, most of our findings are consistent with existing biological knowledge. Based on the personalized driver profiles, we discover two existing and one unreported breast cancer subtypes and uncover their molecular mechanisms. These results intensify our understanding for breast cancer mechanisms, guide therapeutic decisions and assist in the development of targeted anticancer therapies.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Oncogenes , Mutation , Phenotype , Research
7.
Cancers (Basel) ; 15(13)2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37444547

ABSTRACT

Histones play a critical role in chromatin function but are susceptible to mutagenesis. In fact, numerous mutations have been observed in several cancer types, and a few of them have been associated with carcinogenesis. Histones are peculiar, as they are encoded by a large number of genes, and the majority of them are clustered in three regions of the human genome. In addition, their replication and expression are tightly regulated in a cell. Understanding the etiology of cancer mutations in histone genes is impeded by their functional and sequence redundancy, their unusual genomic organization, and the necessity to be rapidly produced during cell division. Here, we collected a large data set of histone gene mutations in cancer and used it to investigate their distribution over 96 human histone genes and 68 different cancer types. This analysis allowed us to delineate the factors influencing the probability of mutation accumulation in histone genes and to detect new histone gene drivers. Although no significant difference in observed mutation rates between different histone types was detected for the majority of cancer types, several cancers demonstrated an excess or depletion of mutations in histone genes. As a consequence, we identified seven new histone genes as potential cancer-specific drivers. Interestingly, mutations were found to be distributed unevenly in several histone genes encoding the same protein, pointing to different factors at play, which are specific to histone function and genomic organization. Our study also elucidated mutational processes operating in genomic regions harboring histone genes, highlighting POLE as a factor of potential interest.

8.
Int J Mol Sci ; 24(7)2023 Mar 29.
Article in English | MEDLINE | ID: mdl-37047418

ABSTRACT

Accurate prediction of the prognoses of cancer patients and identification of prognostic biomarkers are both important for the improved treatment of cancer patients, in addition to enhanced anticancer drugs. Many previous bioinformatic studies have been carried out to achieve this goal; however, there remains room for improvement in terms of accuracy. In this study, we demonstrated that patient-specific cancer driver genes could be used to predict cancer prognoses more accurately. To identify patient-specific cancer driver genes, we first generated patient-specific gene networks before using modified PageRank to generate feature vectors that represented the impacts genes had on the patient-specific gene network. Subsequently, the feature vectors of the good and poor prognosis groups were used to train the deep feedforward network. For the 11 cancer types in the TCGA data, the proposed method showed a significantly better prediction performance than the existing state-of-the-art methods for three cancer types (BRCA, CESC and PAAD), better performance for five cancer types (COAD, ESCA, HNSC, KIRC and STAD), and a similar or slightly worse performance for the remaining three cancer types (BLCA, LIHC and LUAD). Furthermore, the case study for the identified breast cancer and cervical squamous cell carcinoma prognostic genes and their subnetworks included several pathways associated with the progression of breast cancer and cervical squamous cell carcinoma. These results suggested that heterogeneous cancer driver information may be associated with cancer prognosis.


Subject(s)
Breast Neoplasms , Carcinoma, Squamous Cell , Uterine Cervical Neoplasms , Female , Humans , Oncogenes , Breast Neoplasms/genetics , Computational Biology/methods , Carcinoma, Squamous Cell/genetics , Uterine Cervical Neoplasms/genetics
9.
Metabolites ; 13(3)2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36984779

ABSTRACT

Cancer is a heterogeneous disease that is driven by the accumulation of both genetic and nongenetic alterations, so integrating multiomics data and extracting effective information from them is expected to be an effective way to predict cancer driver genes. In this paper, we first generate comprehensive instructive features for each gene from genomic, epigenomic, transcriptomic levels together with protein-protein interaction (PPI)-networks-derived attributes and then propose a novel semisupervised deep graph learning framework GGraphSAGE to predict cancer driver genes according to the impact of the alterations on a biological system. When applied to eight tumor types, experimental results suggest that GGraphSAGE outperforms several state-of-the-art computational methods for driver genes identification. Moreover, it broadens our current understanding of cancer driver genes from multiomics level and identifies driver genes specific to the tumor type rather than pan-cancer. We expect GGraphSAGE to open new avenues in precision medicine and even further predict drivers for other complex diseases.

10.
Comput Biol Med ; 151(Pt A): 106236, 2022 12.
Article in English | MEDLINE | ID: mdl-36370584

ABSTRACT

By taking a new perspective to combine a machine learning method with an evolutionary algorithm, a new hybrid algorithm is developed to predict cancer driver genes. Firstly, inspired by the search strategy with the capability of global search in evolutionary algorithms, a gravitational kernel is proposed to act on the full range of gene features. Constructed by fusing PPI and mutation features, the gravitational kernel is capable to produce repulsion effects. The candidate genes with greater mutation effects and PPI have higher similarity scores. According to repulsion, the similarity score of these promising genes is larger than ordinary genes, which is beneficial to search for these promising genes. Secondly, inspired by the idea of elite populations related to evolutionary algorithms, the concept of vital few is proposed. Targeted at a local scale, it acts on the candidate genes associated with vital few genes. Under attraction effect, these vital few driver genes attract those with similar mutational effects to them, which leads to greater similarity scores. Lastly, the model and parameters are optimized by using an evolutionary algorithm, so as to obtain the optimal model and parameters for cancer driver gene prediction. Herein, a comparison is performed with six other advanced methods of cancer driver gene prediction. According to the experimental results, the method proposed in this study outperforms these six state-of-the-art algorithms on the pan-oncogene dataset.


Subject(s)
Algorithms , Neoplasms , Humans , Oncogenes , Machine Learning , Mutation , Neoplasms/genetics
11.
Iran J Biotechnol ; 20(2): e3066, 2022 Apr.
Article in English | MEDLINE | ID: mdl-36337068

ABSTRACT

Background: Cancer is a group of diseases that have received much attention in biological research because of its high mortality rate and the lack of accurate identification of its root causes. In such studies, researchers usually try to identify cancer driver genes (CDGs) that start cancer in a cell. The majority of the methods that have ever been proposed for the identification of CDGs are based on gene expression data and the concept of mutation in genomic data. Recently, using networking techniques and the concept of influence maximization, some models have been proposed to identify these genes. Objectives: We aimed to construct the cancer transcriptional regulatory network and identify cancer driver genes using a network science approach without the use of mutation and genomic data. Materials and Methods: In this study, we will employ the social influence network theory to identify CDGs in the human gene regulatory network (GRN) that is based on the concept of influence and power of webpages. First, we will create GRN Networks using gene expression data and Existing nodes and edges. Next, we will implement the modified algorithm on GRN networks being studied by weighting the regulatory interaction edges using the influence spread concept. Nodes with the highest ratings will be selected as the CDGs. Results: The results show our proposed method outperforms most of the other computational and network-based methods and show its superiority in identifying CDGs compared to many other methods. In addition, the proposed method can identify many CDGs that are overlooked by all previously published methods. Conclusions: Our study demonstrated that the Google's PageRank algorithm can be utilized and modified as a network-based method for identifying cancer driver gene in transcriptional regulatory network. Furthermore, the proposed method can be considered as a complementary method to the computational-based cancer driver gene identification tools.

12.
Front Genet ; 13: 937948, 2022.
Article in English | MEDLINE | ID: mdl-36017503

ABSTRACT

Hepatocellular carcinoma (HCC) is a highly malignant and heterogeneous tumor with poor prognosis. Cancer driver genes (CDGs) play an important role in the carcinogenesis and progression of HCC. In this study, we comprehensively investigated the expression, mutation, and prognostic significance of 568 CDGs in HCC. A prognostic risk model was constructed based on seven CDGs (CDKN2C, HRAS, IRAK1, LOX, MYCN, NRAS, and PABPC1) and verified to be an independent prognostic factor in both TCGA and ICGC cohorts. The low-score group, which showed better prognosis, had a high proportion of CD8+ T cells and elevated expression of interferon-related signaling pathways. Additionally, we constructed a nomogram to extend the clinical applicability of the prognostic model, which exhibits excellent predictive accuracy for survival. Our study showed the important role of CDGs in HCC and provides a novel prognostic indicator for HCC.

13.
Brief Bioinform ; 23(5)2022 09 20.
Article in English | MEDLINE | ID: mdl-35901472

ABSTRACT

MOTIVATION: Digital pathological analysis is run as the main examination used for cancer diagnosis. Recently, deep learning-driven feature extraction from pathology images is able to detect genetic variations and tumor environment, but few studies focus on differential gene expression in tumor cells. RESULTS: In this paper, we propose a self-supervised contrastive learning framework, HistCode, to infer differential gene expression from whole slide images (WSIs). We leveraged contrastive learning on large-scale unannotated WSIs to derive slide-level histopathological features in latent space, and then transfer it to tumor diagnosis and prediction of differentially expressed cancer driver genes. Our experiments showed that our method outperformed other state-of-the-art models in tumor diagnosis tasks, and also effectively predicted differential gene expression. Interestingly, we found the genes with higher fold change can be more precisely predicted. To intuitively illustrate the ability to extract informative features from pathological images, we spatially visualized the WSIs colored by the attention scores of image tiles. We found that the tumor and necrosis areas were highly consistent with the annotations of experienced pathologists. Moreover, the spatial heatmap generated by lymphocyte-specific gene expression patterns was also consistent with the manually labeled WSIs.


Subject(s)
Neoplasms , Oncogenes , Humans , Machine Learning , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/pathology
14.
Int J Cancer ; 151(12): 2107-2114, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-35860988

ABSTRACT

Cancer precision medicine with biomarker of cancer driver gene (CDG) has been achieved by many small-molecular kinase inhibitors (SMKIs) approved by the US Food and Drug Administration (FDA). Publicly available FDA documents were collected for all SMKI cancer drugs approved between January 2001 and December 2021. Characteristics of indication and pivotal trial were compared. We identified 62 SMKI cancer drugs with 150 indications approved by the FDA between 2001 and 2021. Of these, 55 indications (36.7%) were CDG biomarker-directed. There was a significant increase of 20.5% per year in the number of approved CDG biomarker-directed indications. CDG biomarker-directed indications were associated with significantly higher odds in receiving accelerated approval (odds ratio [OR] = 2.728; 95% CI, 1.246-5.973; P = .012), designating orphan drug (OR = 3.952; 95% CI, 1.758-8.883; P < .001), initial submission of the application (OR = 2.246; 95% CI, 1.063-4.746; P = .034) and in solid cancer (OR = 7.613; 95% CI, 2.958-19.590; P < .001), and were associated with significantly lower odds in using randomized controlled trials (RCTs) (OR = 0.103; 95% CI, 0.032-0.338; P < .001) with less number of entered patients (OR = 0.998; 95% CI, 0.997-1.000; P = .048). The number of CDG biomarker-directed indications in approved SMKIs increased significantly in past two decades, with higher proportion of approvals using special expedited development and approval pathways at the FDA. Further RCTs should be conducted to prove long-term effectiveness and safety.


Subject(s)
Antineoplastic Agents , Neoplasms , United States , Humans , United States Food and Drug Administration , Cross-Sectional Studies , Drug Approval , Antineoplastic Agents/therapeutic use , Biomarkers , Neoplasms/drug therapy , Neoplasms/genetics
15.
BMC Bioinformatics ; 23(1): 230, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35705908

ABSTRACT

Abundant datasets generated from various big science projects on diseases have presented great challenges and opportunities, which contributed to unfolding the complexity of diseases. The discovery of disease-associated molecular networks for each individual plays an important role in personalized therapy and precision treatment of cancer-based on the reference networks. However, there are no effective ways to distinguish the consistency of different reference networks. In this study, we developed a statistical method, i.e. a sample-specific differential network (SSDN), to construct and analyze such networks based on gene expression of a single sample against a reference dataset. We proved that the SSDN is structurally consistent even with different reference datasets if the reference dataset can follow certain conditions. The SSDN also can be used to identify patient-specific disease modules or network biomarkers as well as predict the potential driver genes of a tumor sample.


Subject(s)
Gene Regulatory Networks , Neoplasms , Biomarkers/metabolism , Gene Expression Profiling , Humans , Neoplasms/genetics
16.
Aging (Albany NY) ; 14(5): 2383-2399, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35288483

ABSTRACT

Despite the high prevalence of gastric cancer (GC), molecular biomarkers that can reliably detect GC are yet to be discovered. The present study aimed to establish a robust gene signature based on cancer driver genes (CDGs) that can predict GC prognosis. Transcriptional profiles and clinical data from GC patients were analyzed using univariate Cox regression analysis and the least absolute shrinkage and selection (LASSO)-penalized Cox regression analysis to select optimal prognosis-related genes for modeling. Time-dependent receiver operating characteristic (ROC) and Kaplan-Meier analyses were done to assess the predictive power of this gene signature. A nomogram model for prediction of survival of GC patients was established using the CDG signature and clinical information, and a seven-CDG signature was identified. Risk scores were calculated using this signature, and patients were subsequently divided into high- and low-risk groups; high-risk patients in the training and validation datasets had poorer prognoses than low-risk patients. Cox regression analysis revealed that the CDG signature is an independent prognostic factor for GC. The signature and other clinical features were used to construct a nomogram for predicting overall GC patient survival. Calibration and decision curve analysis showed that the nomogram accurately predicted survival, highlighting its clinical utility. Thus, we established a novel CDG signature and nomogram for predicting GC prognosis, which may facilitate personalized treatment of GC.


Subject(s)
Stomach Neoplasms , Humans , Kaplan-Meier Estimate , Nomograms , Prognosis , ROC Curve , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics
17.
Microorganisms ; 10(2)2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35208856

ABSTRACT

Marek's disease virus (MDV) is the causative agent for Marek's disease (MD), which is characterized by T-cell lymphomas in chickens. While the viral Meq oncogene is necessary for transformation, it is insufficient, as not every bird infected with virulent MDV goes on to develop a gross tumor. Thus, we postulated that the chicken genome contains cancer driver genes; i.e., ones with somatic mutations that promote tumors, as is the case for most human cancers. To test this hypothesis, MD tumors and matching control tissues were sequenced. Using a custom bioinformatics pipeline, 9 of the 22 tumors analyzed contained one or more somatic mutation in Ikaros (IKFZ1), a transcription factor that acts as the master regulator of lymphocyte development. The mutations found were in key Zn-finger DNA-binding domains that also commonly occur in human cancers such as B-cell acute lymphoblastic leukemia (B-ALL). To validate that IKFZ1 was a cancer driver gene, recombinant MDVs that expressed either wild-type or a mutated Ikaros allele were used to infect chickens. As predicted, birds infected with MDV expressing the mutant Ikaros allele had high tumor incidences (~90%), while there were only a few minute tumors (~12%) produced in birds infected with the virus expressing wild-type Ikaros. Thus, in addition to Meq, key somatic mutations in Ikaros or other potential cancer driver genes in the chicken genome are necessary for MDV to induce lymphomas.

18.
Brief Bioinform ; 23(2)2022 03 10.
Article in English | MEDLINE | ID: mdl-35037014

ABSTRACT

Optimal methods could effectively improve the accuracy of predicting and identifying candidate driver genes. Various computational methods based on mutational frequency, network and function approaches have been developed to identify mutation driver genes in cancer genomes. However, a comprehensive evaluation of the performance levels of network-, function- and frequency-based methods is lacking. In the present study, we assessed and compared eight performance criteria for eight network-based, one function-based and three frequency-based algorithms using eight benchmark datasets. Under different conditions, the performance of approaches varied in terms of network, measurement and sample size. The frequency-based driverMAPS and network-based HotNet2 methods showed the best overall performance. Network-based algorithms using protein-protein interaction networks outperformed the function- and the frequency-based approaches. Precision, F1 score and Matthews correlation coefficient were low for most approaches. Thus, most of these algorithms require stringent cutoffs to correctly distinguish driver and non-driver genes. We constructed a website named Cancer Driver Catalog (http://159.226.67.237/sun/cancer_driver/), wherein we integrated the gene scores predicted by the foregoing software programs. This resource provides valuable guidance for cancer researchers and clinical oncologists prioritizing cancer driver gene candidates by using an optimal tool.


Subject(s)
Neoplasms , Oncogenes , Algorithms , Computational Biology/methods , Gene Regulatory Networks , Humans , Mutation , Neoplasms/genetics , Software
19.
Front Immunol ; 12: 728853, 2021.
Article in English | MEDLINE | ID: mdl-35140701

ABSTRACT

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


Subject(s)
Adenocarcinoma of Lung/immunology , Autoantibodies/immunology , Carcinoembryonic Antigen/immunology , Immunoglobulin M/immunology , Lung Neoplasms/immunology , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/immunology , Biomarkers, Tumor/immunology , Carcinoma, Squamous Cell/immunology , Early Detection of Cancer/methods , Female , Humans , Male , Middle Aged , Oncogenes/immunology , ROC Curve
20.
J Biomed Inform ; 114: 103661, 2021 02.
Article in English | MEDLINE | ID: mdl-33326867

ABSTRACT

Cancer is among the diseases causing death, in which, cells uncontrollably grow and reproduce beyond the cell regulatory mechanism. In this disease, some genes are initiators of abnormalities and then transmit them to other genes through protein interactions. Accordingly, these genes are known as cancer driver genes (CDGs). In this regard, several methods have been previously developed for identifying cancer driver genes. Most of these methods are computational-based, which use the concept of mutation to predict CDGs. In this research, a method has been proposed for identifying CDGs in the transcription regulatory network using the concept of influence diffusion and by modifying the Hyperlink-Induced Topic Search algorithm based on the diffusion concept. Due to the type of these networks and the processes of abnormality progression in cells and the formation of cancerous tumors, high-influence genes can be the most likely considered as the driver genes. Therefore, we can use the influence diffusion concept as an acceptable theory to identify these genes. Recently, a method has been proposed to detect CDGs with the concept of the influence maximization. One of the challenges in these types of networks is finding the power of regulatory interaction between genes. Moreover, we have proposed a novel method to calculate the weight of regulatory interactions, based on the concept of diffusion. The performance of the proposed method was compared with other seventeen computational and network tools. Correspondingly, three cancer types were used as benchmarks as follows: breast invasive carcinoma (BRCA), Colon adenocarcinoma (COAD), and lung squamous cell carcinoma (LUSC). In addition, to determine the accuracy of the detected drivers using each method, CGC (Cancer Gene Census) and Mut-driver gene lists were utilized as gold standard. The results show that GenHITS performs better compared to the most of the other computational and network methods. Besides, it is also able to identify genes that have been identified by none of the other methods yet.


Subject(s)
Breast Neoplasms , Oncogenes , Algorithms , Breast Neoplasms/genetics , Female , Gene Regulatory Networks , Humans , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL