Skip to main content
/v1/supplement/title

Identification of gene profiles related to the development of oral cancer using a deep learning technique

Abstract

Background

Oral cancer (OC) is a debilitating disease that can affect the quality of life of these patients adversely. Oral premalignant lesion patients have a high risk of developing OC. Therefore, identifying robust survival subgroups among them may significantly improve patient therapy and care. This study aimed to identify prognostic biomarkers that predict the time-to-development of OC and survival stratification for patients using state-of-the-art machine learning and deep learning.

Methods

Gene expression profiles (29,096 probes) related to 86 patients from the GSE26549 dataset from the GEO repository were used. An autoencoder deep learning neural network model was used to extract features. We also used a univariate Cox regression model to select significant features obtained from the deep learning method (P < 0.05). High-risk and low-risk groups were then identified using a hierarchical clustering technique based on 100 encoded features (the number of units of the encoding layer, i.e., bottleneck of the network) from autoencoder and selected by Cox proportional hazards model and a supervised random forest (RF) classifier was used to identify gene profiles related to subtypes of OC from the original 29,096 probes.

Results

Among 100 encoded features extracted by autoencoder, seventy features were significantly related to time-to-OC-development, based on the univariate Cox model, which was used as the inputs for the clustering of patients. Two survival risk groups were identified (P value of log-rank test = 0.003) and were used as the labels for supervised classification. The overall accuracy of the RF classifier was 0.916 over the test set, yielded 21 top genes (FUT8-DDR2-ATM-CD247-ETS1-ZEB2-COL5A2-GMAP7-CDH1-COL11A2-COL3A1-AHR-COL2A1-CHORDC1-PTP4A3-COL1A2-CCR2-PDGFRB-COL1A1-FERMT2-PIK3CB) associated with time to developing OC, selected among the original 29,096 probes.

Conclusions

Using deep learning, our study identified prominent transcriptional biomarkers in determining high-risk patients for developing oral cancer, which may be prognostic as significant targets for OC therapy. The identified genes may serve as potential targets for oral cancer chemoprevention. Additional validation of these biomarkers in experimental prospective and retrospective studies will launch them in OC clinics.

Peer Review reports

Introduction

Oral cancer is among the 10th most prevalent cancer types among men and the 12th most prevalent cancer among women worldwide [1]. Oral cancer is mainly observed in the tongue, nevertheless, it can occur on many sites including the gingiva, palate, lips as well as the floor of the mouth, cheeks, and the area behind the wisdom teeth [2]. Oral squamous cell carcinoma (OSCC) accounts for over 90% of head and neck cancers (with ~ 450,000 new cases annually) [3, 4]. The incidence and deaths due to oral cancer deaths vary across the world, with the highest in developing countries [5]. Studies have shown that regardless of the advancements in screening approaches and pharmacological treatments, the incidence and mortality rates of oral cancer are practically increasing [6, 7]. Detecting oral cancer in its early stages, as well as timely treatment of the disease, are considered the most efficient ways of controlling the mortality rate [1]; nevertheless, most the oral tumors are diagnosed at an advanced stage which reduces the patients’ survival [8]. There are various treatments for OSCC, including surgery, radiotherapy, and adjunct chemotherapy (sometimes in combination), depending on the stage of the disease. However, OSCC has a poor prognosis, so the five-year overall survival rate of OSCC is less than 50% (ranges from 15 to 60%) [9, 10], depending on the severity of the disease [11].

The role of several factors including smoking, age, alcohol consumption, infections sustained by human papillomaviruses (HPVs), Epstein-Barr virus (EBV), or Candida albicans in developing OSCC, has been well-established [12,13,14]. Several studies have proposed some biomarkers for diagnosing oral cancer lesions with somehow low sensitivity/specificity for effective diagnosing of all oral tumors [15]. However, only 15% of all pharmaceutical agents have demonstrated “sufficient safety and potency to gain any sort of regulatory consent” [16]. Moreover, the biomarkers associated with the time-to-OSCC development (patient survival as the objective) that can be used in the prognosis of OSCC have not been fully understood and remained obscure. This indicates the deficiencies in the understanding of cancer complexity and highlights the importance of the identification of new prognostic biomarkers to obtain information for monitoring patients effectively and managing the treatment process [16]. Therefore, discovering biomarkers based on gene profiles data that are involved in the development of OSCC and improvement of survival prediction using state-of-the-art models is much needed in patients with oral preneoplastic lesions. This provides models that can mimic “the diversity of human tumor biology in a preclinical arrangement” efficiently [16], which would help an improved prognosis of OSCC and early treatment.

During the past decades, a vast majority of the literature has considered expert models in ultra-high dimensional feature spaces extensively. Among them, deep learning (DL), which is an advanced computer-aided technique, has gained much attention in the medical field [17], and it has been shown to have a vital role in detecting and diagnosing different types of cancer as well as prognosis of a disease. Autoencoders are types of DL usually used for unsupervised objectives, and recently, they have received much attention for unsupervised feature extraction in survival analysis. Studies have established autoencoder as an efficient approach to produce features related to some clinical outcomes like time-to-event response [18, 19] and have utilized autoencoders for analyzing genomics and expression data in other cancers for unsupervised feature selection as inputs of survival analysis [20, 21].

DL has been widely used in various cancers to predict the survival of patients. For example, Zhang et al. conducted a study based on the features obtained by an autoencoder algorithm to identify prognostic subtypes of high-risk neuroblastoma using multi-omics data. They showed that the autoencoder outperformed other methods like the principal component method in terms of prognosis [22]. Takahashi et al. have used an autoencoder to predict the survival of patients with lung cancer using omics data. They identified survival-associated subtypes in non-small cell lung cancer (longer and shorter-surviving groups) [23]. Chaudhary et al. utilized an autoencoder in analyzing liver cancer data and demonstrated that the DL used provides robust prognostic subtypes in liver cancer using omics data. Moreover, some attempts have been made to apply DL in the diagnosis and prognosis of OSCC. Parallel to our study, Li et al. have used an autoencoder to identify molecular subtypes of OSCC focusing on immunosuppression genes. However, they have used a different pipeline. Also, Shams and Htike have used deep neural networks using feed-forward with backpropagation design to diagnose, and predict oral cancer versus healthy controls based on gene expression profiling [24]. However, the former limited their analysis to the immunosuppressive genes and the latter handled a classification problem.

Yet, not all aspects of the time-to-development of OSCC have been fully understood. Therefore, this study used an unsupervised autoencoder framework to build a model for predicting the prognosis of OSCC patients to provide a prognostic stratification for the survival of the patients and to identify potential effective biomarkers related to the prognosis of oral cancer in patients with the oral preneoplastic lesions.

Methods

Data source and preprocessing

GSE26549 dataset from the GEO repository related to oral cancer tissue transcriptome (generated using the Affymetrix transcript version (microarray) with platform ID GPL6244) was used. This dataset consisted of preprocessed expression data of 86 oral preneoplastic lesion (OPL) patients, and thirty-five out of the 86 patients developed oral cancer [25]. Survival time was defined as the time to develop the oral preneoplastic lesions to oral cancer in patients. The individuals with oral preneoplastic lesions not develop oral cancer were considered as censor. The quantile normalization (QN) procedure was utilized using “bestNormalize” R package.

Feature extraction using DL framework

Here, we used the DL computational framework on gene expression profiles related to developing OSCC in patients with the oral preneoplastic lesions. An autoencoder framework was selected as the implementation of DL for feature extraction. The philosophy of autoencoder is similar to the principal component analysis, where linear combinations of the original variables are constructed. Autoencoders receive the gene expression profiles as the inputs and reconstruct the original input by combining some nonlinear functions. These combinations are then used as new features and can be used as inputs for further analysis instead of the original variables [19]. The preprocessed gene expression profiles related to 86 samples were used as the input for the autoencoder framework. An autoencoder is a feed-forward, nonrecurrent neural network that learns through unsupervised learning [26], and is trained to reconstruct the original input to its output. Let us consider \(x=\left({x}_{1}.\dots .{x}_{n}\right)\) as the input vector of dimension n of the input layer of an autoencoder. So, the autoencoder aims to reconstruct the x vector by an x' vector (dimension n). This is done by providing successive transformations of x in several hidden layers. In this study, for the ith layer, the rectified linear activation function or ReLU activation function was used [27] between input layer x and output layer y, i.e.,

$$y={f}_{i}(x)=\mathrm{ReLU}\left({W}_{i}x+{b}_{i}\right)$$

where x is a vector of size d and y is a vector of size p. Also, \({W}_{i}\) stands for a \(p\times d\) weight matrix and \({b}_{i}\) stands for the intercept vector with size p. For an autoencoder with k layers, x' is then given by:

$${x}^{\mathrm{^{\prime}}}={F}_{1\to k}(x)={f}_{1}^{\circ }{\dots }^{\circ }{f}_{k-1}^{\circ }{f}_{k}(x)$$

where \({f}_{k-1}{ }^{\circ }{f}_{k}(x)={f}_{k-1}\left({f}_{k}(x)\right)\) is the composed function of \({f}_{k-1}\) with \({f}_{k}\).

An autoencoder is trained so that different weight vectors of \({W}_{i}\) are obtained to optimize (minimization problem) a specific objective function like mean squared error (MSE), measuring the error between the input \(x\) and the output \({x}^{\mathrm{^{\prime}}}\) as follows:

$$MSE(x\cdot {x}^{\mathrm{^{\prime}}})=\frac{1}{N}\sum_{i=1}^{N}{\left({x}_{i}-{{x}^{\mathrm{^{\prime}}}}_{i}\right)}^{2}$$

Also, an \(L1\) penalty (say, \({\alpha }_{w}\)) on the weight vector of \({W}_{i}\) and an \(L2\) penalty (say, \({\alpha }_{a}\)) on the activities of the nodes, \({F}_{1\to k}\left(x\right)\) was added to the objective function to control overfitting as follows [28]:

$$L\left(x\cdot { x}^{{{\prime}}}\right)=MSE(x\cdot {x}^{{{\prime}}})+\sum_{i=1}^{N} \left({\alpha }_{w}{\parallel{W}_{i}\parallel }_{1}+{\alpha }_{a}{\parallel {F}_{1\to i}\left(x\right)\parallel}_{2}^{2}\right)$$

In this study, the Python Keras package (https://github.com/fchollet/keras) was utilized to build an autoencoder consisting of three hidden layers (500, 100 and 500 nodes, respectively). The bottleneck layer of the autoencoder was used to extract new features from the gene profiles of oral cancer patients. Finally, the gradient descent approach [29] with 140 epochs (iterations) and 50% dropout were utilized for training the autoencoder as the learning algorithm. Each instance of training data is processed once by the learning algorithm during one epoch. Both regularization parameters of L1 and L2 were obtained 0.0001 through cross-validation. A number of 140 epochs were used.

Hierarchical clustering and feature selection

The extracted features from the autoencoder were examined through the univariate Cox proportional hazards (Cox-PH) model [30] to select significant features (\(P<0.05\)). Then, the significant features were used to cluster the patients through the hierarchical clustering algorithm [31].

Identifying low and high-risk groups

The Kaplan–Meier curve and the log-rank test were used to identify survival groups (high-risk and low-risk groups). According to Kaplan Meier's curve, the patients with lower median survival were considered as the high-risk survival patients, and the other group was regarded as the low-risk group [32]. Also, the prognostic index (\(\beta^{\prime}X\)) was calculated for the external validation set, where \(\beta^{\prime}\) stands for the regression coefficients obtained from a multivariate Cox regression model and X indicates the matrix of selected genes.

Supervised random forest classifier

The survival groups identified in the previous step were considered new labels for the patients. A supervised random forest classifier was created to choose the risk-related genes. RF, introduced by [33], constructs many classification/regression trees through randomly selected training datasets and random subsets of predictors for predicting outcomes. The final prediction of the outcome is calculated by aggregating the predictions provided by each tree. So, higher accuracy is achieved by RF compared to a single decision tree model [34]. Also, RF provides variable importance criterion for variable selection. In this study, variable importance was used to select key genes [35]. The tenfold cross-validation technique was used to tune the parameters of the RF. Finally, the cut-off point of 0.002 was used as a criterion for gene selection.

Gene Ontology (GO) and KEGG pathway enrichment analysis

The Database for Annotation Visualization and Integrated Discovery (DAVID) program was used for GO and KEGG pathway [36,37,38] enrichment analysis for 21 selected genes by the RF method to see if they have a role in biological process, cellular component, etc. The Benjamini adjusted P-value less than 0.05 was considered statistically significant. Gene ontology provides information that helps to computationally analyze and achieve knowledge about gene functions determined by large-scale molecular biology approaches and genetic experiments [2]. Pathways associated with genes are also provided in gene ontology.

Protein–protein interaction (PPI) network analysis

The PPI network was constructed using the “Search Tool for the Retrieval of Interacting Genes” (STRING) for 21 selected genes. A confidence score of 0.4 was considered as a threshold for network construction. Afterward, the PPI network was visualized and analyzed by Cytoscape software (V 3.8.2).

Results

Thirty-five out of 86 patients developed oral cancer. The mean and median follow-up time of the patients were 9.01 and 10.7 years (min = 0.18 and max = 14.34 years), respectively. One-, three- and five-year survival rates of the patients were 88%, 73%, and 65%, respectively.

The information on gene expression profiles related to 86 patients with oral preneoplastic lesions was used as input features of the autoencoder, a DL framework. Figure 1 illustrates the architecture of the autoencoder (a) and the loss values (MSE) versus the epochs (b). The activity of the 100 nodes from the bottleneck hidden layer was extracted as new features. Seventy features out of the 100 new features were statistically significant using univariate Cox-PH regression (P < 0.05), and they were shown to be associated with the survival of the patients. These 70 features were subjected to hierarchical clustering, with cluster number K ranging from 2 to 6. Considering the silhouette index, the number of 2 (k = 2) clusters was the optimum. Table 1 shows the characteristics of each group identified using clustering based on 70 features from DL. The median survival for group 1 was ~ 5 years and ~ 10 years. Furthermore the survival analysis on the entire data (86 patients) showed that the survival curves in the two identified clusters (Fig. 2) were statistically different (log-rank test P = 0.003). We also conducted the penalized principal component analysis as an alternative to the DL. Using univariate Cox regression, 20 out of 100 principal components were significant (P < 0.05). The same strategy was used to identify two groups, but the difference between the two survival curves using this method was not statistically significant (P = 0.171). Thus, the two classes were considered labels for the subsequent supervised RF classifier.

Fig. 1
figure 1

a Architecture of the autoencoder, and b loss function values over epochs

Table 1 Survival information of two identified groups
Fig. 2
figure 2

Kaplan Meier curve for two subgroup of survival time

The RF classifier was trained (sensitivity = 0.814, specificity = 0.966, and total accuracy = 0.916 over the 30% of data as a testing set), and gene profiles related to the survival risk groups were selected based on variable importance greater than 0.002. Table 2 shows the variable importance of 21 top-rank genes with variable importance greater than 0.002 and their over/under expression status in high-risk patients. Also, Fig. 3 illustrates the heatmap of the 21 selected genes.

Table 2 Top genes identified by random forest method through variable importance (VIMP)
Fig. 3
figure 3

Heat-map of the 21 selected genes using random forest related two identified survival groups

Gene Ontology (GO) and KEGG pathway enrichment analysis

Summary of the top GO results and KEGG pathways were illustrated in Fig. 4. The results of GO enrichment analysis indicated that collagen fibril organization, extracellular matrix organization, skeletal system development, cellular response to amino acid stimulus, regulation of immune response, transforming growth factor beta receptor signaling pathway, platelet activation, tissue homeostasis, skin development, blood vessel development, and chondrocyte differentiation terms were significantly enriched in biological process (BP). Collagen trimer, extracellular matrix, endoplasmic reticulum lumen, collagen type I trimer, extracellular region, and Golgi apparatus were significantly enriched in cellular component (CC). Platelet-derived growth factor binding, extracellular matrix structural constituent conferring tensile strength, extracellular matrix structural constituent, SMAD binding, and identical protein binding terms were significantly enriched in molecular function (MF).

Fig. 4
figure 4

Summary of the top GO results and KEGG pathways

The KEGG pathway analysis indicated that the protein digestion and absorption, human papillomavirus infection, focal adhesion, AGE-RAGE signaling pathway in diabetic complications, amoebiasis, platelet activation, and relaxin signaling pathway terms were enriched for 21 selected genes (Fig. 4).

PPI network analysis

The constructed PPI network was composed of 15 nodes and 38 edges. In order to find the hub genes, including in the pathogenesis of oral cancer, the constructed PPI network was evaluated with the CytoHubba package under Cytoscape software. The top 10 genes were identified using the four methods, including degree, MNC, EPC, and EcCentricity. After depicting the Venn diagram (Fig. 5), eight common genes including PDGFRB, COL1A2, CDH1, DDR2, COL3A1, COL2A1, COL1A1, and COL5A2 were selected as hub genes (Fig. 6).

Fig. 5
figure 5

The overlap between the top predicted target genes, ranked by MNC, MCC, and degree, is illustrated in a Venn diagram

Fig. 6
figure 6

The PPI network of identified genes, formed by using Cytoscape software. Proteins are represented by nodes, and interactions between two proteins are described by edges

In silico validation of selected genes

An in silico validation was conducted for the selected probes using two public data sets with series accession numbers of GSE9844 and GSE41613 on oral cancer, so that we predicted the outcome of independent samples related to external data sets based on the selected genes from the previous steps using the utilized method in the original data set (GSE26549). The first dataset included gene expression profiles of 26 microdissected OTSCC tissues and 12 matching normal tissue samples [39]. We applied the RF method for the classification of the oral cancer patients and healthy controls. About 70% of the data was considered as a training set, and the rest of them were used to test the method. On the training data set, a three-fold cross-validation strategy was used to tune the parameters. Figure 7a depicts the ROC curve along with the AUC for the testing set in the in silico validation data set using the selected genes in Table 2. According to the results, the AUC was 1.000, indicating that the identified genes can successfully predict oral cancer development and can be used for the prognosis of the patients.

Fig. 7
figure 7

a ROC curve related to the prediction of oral cancer patients and healthy controls in in silico validation data set (GSE9844); b prediction error curve in predicting survival of oral cancer patients over GSE41613 data set as in silico validation; c Kaplan-Mayer curves of survival subgroups identified using selected genes over GSE41613 data set as in silico validation

The second data set included the survival time of 97 oral cancer patients. The prediction error curve, based on a model including selected genes in the previous steps, was provided in Fig. 7b indicating that the selected genes were potentially informative in predicting the survival of the patients with oral cancer. Also, we calculated the prognostic index (\(\beta^{\prime}X\), where \(\beta^{\prime}\) stands for the regression coefficients obtained from a multivariate Cox regression model, and X indicates the matrix of selected genes). So, the patients were divided into two risk groups. Figure 7c illustrates the Kaplan–Meier survival curves for the two groups. The log-rank test showed a statistical difference between the curves (Chisq = 8.1 on 1 degree of freedom, P = 0.004).

Discussion

According to the findings of the present study, FUT8 (Fucosyltransferase 8) was the first top gene identified by the algorithm used (DL and RF). FUT8 is a protein-coding gene that encodes an enzyme belonging to the family of fucosyltransferases involved in many pathological/physiological activities such as tumor metastasis and inflammation) [40, 41], and regulating the fucosylation of O-glycans and N-glycans [40]. Results of the present study indicated overexpression of FUT8 in identified high-risk patients compared to the low-risk group, which was in concordance with other studies [42]. Studies have shown increasing mRNA levels of FUT8 and core glycoprotein in tumor tissues of oral cancer patients compared with normal oral epithelial/oesophageal tissue [42, 43]. “FUT8 plays an anti-radiation-driven role in ESCC by core fucosylation of CD147, and it can be used as a marker to predict the radiotherapy response of ESCC patients” [40, 44]. According to the findings, DDR2 (Discoidin domain receptor 2) was the second top rank gene identified by RF, showing overexpression in high-risk patients. This finding was in concordance with the results of other studies [45, 46]. DDR2 is a receptor tyrosine kinase (RTK), and it has been shown to be activated through fibrillar collagens [46] and involved in cell behaviors of different types of cancer, including VEGF expression, differentiation, tumor angiogenesis, invasion, and metastatic potential of HNSCC cell lines [46]. DDR2 has been well-established to be activated through binding with collagens. Then, a series of intracellular pathways of p38, JNK, ERK1/2, Notch-1, and NF-κB are activated [47, 48]. Several studies have shown the regulatory functions of DDR2 factor in different types of cancers, including lung carcinoma [49]. Ataxia telangiectasia mutated (ATM) was the third top rank gene identified as an important gene in determining high-risk patients. It was shown that it is over-expressed in high-risk survival group patients. This finding was consistent with the results of other studies [50]. ATM encodes a vital cell cycle checkpoint (CCK) kinase protein belonging to the PI3/PI4-kinase family that functions as a regulator of various downstream proteins, including “tumor suppressor proteins p53 and BRCA1, checkpoint kinase CHK2, checkpoint proteins RAD17 and RAD9, and DNA repair protein NBS1”. This protein is thought to be one of the two master controllers of CCK signaling pathways essential in cell response to DNA damage and genome stability [51].

The protein encoded by PDGFRB as a plasma membrane receptor belongs to the platelet-derived growth factor family. The binding of PDGF ligands to this receptor leads to dimerization and activation of downstream signaling pathways having a role in the regulation of motility and proliferation, differentiation, and survival of cells [52]. Lin et al., in their study, indicated that PDGFRB expression level was associated with poor prognosis and lymph node metastasis of OSCC [53]. E-cadherin protein encoded with CDH1 belongs to the cadherin protein family. This transmembrane glycoprotein, which regulates cell adhesion, is a tumor suppressor protein [54]. Pannone et al., in their study, showed that the expression level of CDH1 decreases in oral tumors in mRNA and protein levels. Moreover, the expression level of E-cadherin had a reverse correlation with tumor grade and prognosis of patients [55].

The protein encoded with COL1A2 and COL1A1 is the building block of type I collagen. The expression level of COL1A2 is dysregulated in several tumors such as malignant melanoma head and neck ovarian pancreatic, and bladder cancer [56, 57]. COL3A1 encoded the polypeptide chain which was the building block of type III collagen. The expression of this gene has an essential role in the proliferation and migration of tumor cells and is dysregulated in several malignancies, such as ovarian cancer and brain tumor [58, 59]. Collagen, type II, alpha 1 peptide encoded with COL2A1 form the homotrimer Type II collagen [60]. Tarpey et al., in a study, showed that in Chondrosarcoma, hyper-mutability of COL2A1 is common [61]. Moreover, Ganapathi et al., in a study, indicated that the expression level of this gene is associated with the prognosis of high-grade serous ovarian cancer [62]. Type V collagen consists of peptides encoded with COL5A2. This gene has an essential role in the regulation of angiogenesis and metastasis of several tumors such as osteosarcoma, colorectal cancer, gastric cancer, and breast cancer [63, 64].

The results of GO enrichment analysis indicated that genes identified in this study representing biological pathways were significantly enriched in relation to cancers. These findings were in agreement with similar studies, so that cancer-related terms such as collagen fibril organization, extracellular matrix organization [65], cellular response to amino acid stimulus, platelet activation [66], tissue homeostasis [67], regulation of immune response, skin development [68], platelet-derived growth factor binding, extracellular matrix structural constituent, SMAD binding [66], extracellular matrix [65], collagen trimmer, and endoplasmic reticulum lumen [66] were enriched for 21 selected genes by the RF method. Moreover KEGG pathway enrichment analysis indicated that cancer related pathways such as protein digestion and absorption, platelet activation [65], focal adhesion, Amoebiasis [66], human papillomavirus infection [69], AGE-RAGE signaling pathway in diabetic complications [70], and relaxin signaling pathway [71] were significantly enriched which was in agreement with the results of other studies.

In the present study, we sued a univariate Cox regression model as the multivariate regression could not be applied here due to a large number of unsupervised extracted features (> 100) compared to the sample size (n = 86). It is suggested to use other methods of screening selected features, like penalized Cox regression model with different penalties to choose a subset of features among the pool of features, and to conduct simulation studies to see which variable selection method works better.

Conclusion

This study identified eight hub genes, including PDGFRB, COL1A2, CDH1, DDR2, COL3A1, COL2A1, COL1A1, and COL5A2, that may have a role in development of oral cancer. Further experimental investigations are required in order to well-understand and to validate the pathogenic role of these genes in oral cancer.

Availability of data and materials

The datasets is publically available on https://www.ncbi.nlm.nih.gov/geo/. All analyzed during the current study are available from the corresponding author on request.

References

  1. Glick M. Burket's oral medicine. 2015: PMPH USA.

  2. Ariya S, James A, Joseph B. Computational analysis of oral cancer gene expression profile and identification of MiRNAs and their regulatory hub genes. J Complement Med Res. 2020;11(3):154–9.

    Article  CAS  Google Scholar 

  3. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, Znaor A, Bray F. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144(8):1941–53.

    Article  CAS  PubMed  Google Scholar 

  4. Leemans CR, Snijders PJ, Brakenhoff RH. The molecular landscape of head and neck cancer. Nat Rev Cancer. 2018;18(5):269–82.

    Article  CAS  PubMed  Google Scholar 

  5. Mosaddad SA, Beigi K, Doroodizadeh T, Haghnegahdar M, Golfeshan F, Ranjbar R, Tebyanian H. Therapeutic applications of herbal/synthetic/bio-drug in oral cancer: An update. Eur J Pharmacol. 2021;890: 173657.

    Article  CAS  PubMed  Google Scholar 

  6. Falzone L, Salomone S, Libra M. Evolution of cancer pharmacological treatments at the turn of the third millennium. Front Pharmacol. 2018: 1300.

  7. Gupta N, Gupta R, Acharya AK, Patthi B, Goud V, Reddy S, Garg A, Singla A. Changing trends in oral cancer-a global scenario. Nepal J Epidemiol. 2016;6(4):613.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Grafton-Clarke C, Chen KW, Wilcock J. Diagnosis and referral delays in primary care for oral squamous cell cancer: a systematic review. Br J Gen Pract. 2019;69(679):e112–26.

    Article  PubMed  Google Scholar 

  9. Pruegsanusak K, Peeravut S, Leelamanit V, Sinkijcharoenchai W, Jongsatitpaiboon J, Phungrassami T, Chuchart K, Thongsuksai P. Survival and prognostic factors of different sites of head and neck cancer: an analysis from Thailand. Asian Pac J Cancer Prev. 2012;13(3):885–90.

    Article  PubMed  Google Scholar 

  10. Chen T-C, Hsu C-W, Lou P-J, Ko J-Y, Yang T-L, Chen C-N, Chang Y-L, Wang C-P. The clinical predictive factors for subsequent distant metastasis in patients with locoregionally advanced oral squamous cell carcinoma. Oral Oncol. 2013;49(4):367–73.

    Article  PubMed  Google Scholar 

  11. Guo H, Li C, Su X, Huang X. A five-mRNA expression signature to predict survival in oral squamous cell carcinoma by integrated bioinformatic analyses. Genet Test Mol Biomarkers. 2021;25(8):517–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yete S, D’Souza W, Saranath D. High-risk human papillomavirus in oral cancer: clinical implications. Oncology. 2018;94(3):133–41.

    Article  PubMed  Google Scholar 

  13. She Y, Nong X, Zhang M, Wang M. Correction: Epstein-Barr virus infection and oral squamous cell carcinoma risk: a meta-analysis. PLoS ONE. 2019;14(6): e0217659.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Mohd Bakri M, Mohd Hussaini H, Rachel Holmes A, David Cannon R, Mary Rich A. Revisiting the association between candidal infection and carcinoma, particularly oral squamous cell carcinoma. J Oral Microbiol. 2010;2(1):5780.

    Article  Google Scholar 

  15. Falzone L, Lupo G, La Rosa GRM, Crimi S, Anfuso CD, Salemi R, Rapisarda E, Libra M, Candido S. Identification of novel MicroRNAs and their diagnostic and prognostic significance in oral cancer. Cancers. 2019;11(5):610.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wangmo C, Charoen N, Jantharapattana K, Dechaphunkul A, Thongsuksai P. Epithelial–mesenchymal transition predicts survival in oral squamous cell carcinoma. Pathol Oncol Res. 2020;26(3):1511–8.

    Article  CAS  PubMed  Google Scholar 

  17. Al-Ma’aitah M, AlZubi AA. Enhanced computational model for gravitational search optimized echo state neural networks based oral cancer detection. J Med Syst. 2018;42(11):1–7.

    Article  Google Scholar 

  18. Tan J, Ung M, Cheng C, Greene CS. Unsupervised feature construction and knowledge extraction from genome-wide assays of breast cancer with denoising autoencoders. In Pacific symposium on biocomputing co-chairs. 2014. World Scientific.

  19. Chaudhary K, Poirion OB, Lu L, Garmire LX. Deep learning–based multi-omics integration robustly predicts survival in liver cancer. Clin Cancer Res. 2018;24(6):1248–59.

    Article  CAS  PubMed  Google Scholar 

  20. Chen L, Cai C, Chen V, Lu X. Learning a hierarchical representation of the yeast transcriptomic machinery using an autoencoder model. In BMC bioinformatics. 2016. BioMed Central.

  21. Khalili M, Alavi MH, Khodakarim S, Ahadi B, Hamidpour M. Prediction of the thromboembolic syndrome: an application of artificial neural networks in gene expression data analysis. 2016.

  22. Zhannlg L, Lv C, Jin Y, Cheng G, Fu Y, Yuan D, Tao Y, Guo Y, Ni X, Shi T. Deep learning-based multi-omics data integration reveals two prognostic subtypes in high-risk neuroblastoma. Front Genet. 2018;9:477.

    Article  Google Scholar 

  23. Takahashi S, Asada K, Takasawa K, Shimoyama R, Sakai A, Bolatkan A, Shinkai N, Kobayashi K, Komatsu M, Kaneko S. Predicting deep learning based multi-omics parallel integration survival subtypes in lung cancer using reverse phase protein array data. Biomolecules. 2020;10(10):1460.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Shams WK, Htike ZZ. Oral cancer prediction using gene expression profiling and machine learning. Int J Appl Eng Res. 2017;12(15).

  25. Saintigny P, Zhang L, Fan YH, El-Naggar AK, Papadimitrakopoulou VA, Feng L, Lee JJ, Kim ES, Ki Hong W, Mao L. Gene expression profiling predicts the development of oral cancer. Cancer Prev Res (Phila). 2011;4(2):218–29.

    Article  CAS  PubMed  Google Scholar 

  26. Bengio Y, Learning deep architectures for AI. 2009: Now Publishers Inc.

  27. Schmidt-Hieber J. Nonparametric regression using deep neural networks with ReLU activation function. Ann Stat. 2020;48(4):1875–97.

    Google Scholar 

  28. Odaibo S. Tutorial: Deriving the standard variational autoencoder (VAE) loss function. 2019. arXiv:1907.08956.

  29. Ruder S. An overview of gradient descent optimization algorithms. 2016. arXiv:1609.04747.

  30. Martínez-Camblor P, Mackenzie T, Staiger DO, Goodney PP, O’Malley AJ. Adjusting for bias introduced by instrumental variable estimation in the Cox proportional hazards model. Biostatistics. 2019;20(1):80–96.

    Article  PubMed  Google Scholar 

  31. Murtagh F, Contreras P. Methods of hierarchical clustering. 2011. arXiv:1105.0121.

  32. Goel MK, Khanna P, Kishore J. Understanding survival analysis: Kaplan-Meier estimate. International journal of Ayurveda research. 2010;1(4):274.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Breiman L. Random forests. Mach Learn. 2001;45(1):5–32.

    Article  Google Scholar 

  34. Speiser JL, Miller ME, Tooze J, Ip E. A comparison of random forest variable selection methods for classification prediction modeling. Expert Syst Appl. 2019;134:93–101.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Biau G, Scornet E. A random forest guided tour. TEST. 2016;25(2):197–227.

    Article  Google Scholar 

  36. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kanehisa M. Toward understanding the origin and evolution of cellular organisms. Protein Sci. 2019;28(11):1947–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kanehisa M, Furumichi M, Sato Y, Kawashima M, Ishiguro-Watanabe M. KEGG for taxonomy-based analysis of pathways and genomes. Nucleic Acids Res. 2022.

  39. Ye H, Yu T, Temam S, Ziober BL, Wang J, Schwartz JL, Mao L, Wong DT, Zhou X. Transcriptomic dissection of tongue squamous cell carcinoma. BMC Genom. 2008;9(1):1–11.

    Article  Google Scholar 

  40. Liao C, An J, Yi S, Tan Z, Wang H, Li H, Guan X, Liu J, Wang Q. FUT8 and protein core fucosylation in tumours: from diagnosis to treatment. J Cancer. 2021;12(13):4109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Tu Z, Lin Y-N, Lin C-H. Development of fucosyltransferase and fucosidase inhibitors. Chem Soc Rev. 2013;42(10):4459–75.

    Article  CAS  PubMed  Google Scholar 

  42. Chang S-C, Lin W-L, Chang Y-F, Lee C-T, Wu J-S, Hsu P-H, Chang C-F. Glycoproteomic identification of novel plasma biomarkers for oral cancer. J Food Drug Anal. 2019;27(2):483–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sadeghzadeh Z, Khosravi A, Jazi MS, Asadi J. Upregulation of Fucosyltransferase 3, 8 and protein O-Fucosyltransferase 1, 2 genes in esophageal cancer stem-like cells (CSLCs). Glycoconj J. 2020;37(3):319–27.

    Article  CAS  PubMed  Google Scholar 

  44. Shen L, Xia M, Deng X, Ke Q, Zhang C, Peng F, Dong X, Luo Z. A lectin-based glycomic approach identifies FUT8 as a driver of radioresistance in oesophageal squamous cell carcinoma. Cell Oncol. 2020;43(4):695–707.

    Article  CAS  Google Scholar 

  45. Velmurugan BK, Chang W-H, Chung C-M, Yeh C-M, Lee C-H, Yeh K-T, Lin S-H. DDR2 overexpression in oral squamous cell carcinoma is associated to lymph node metastasis. Cancer Biomark. 2018;22(4):747–53.

    Article  CAS  PubMed  Google Scholar 

  46. Xu J, Lu W, Zhang S, Zhu C, Ren T, Zhu T, Zhao H, Liu Y, Su J. Overexpression of DDR2 contributes to cell invasion and migration in head and neck squamous cell carcinoma. Cancer Biol Ther. 2014;15(5):612–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Vogel W, Gish GD, Alves F, Pawson T. The discoidin domain receptor tyrosine kinases are activated by collagen. Mol Cell. 1997;1(1):13–23.

    Article  CAS  PubMed  Google Scholar 

  48. Croissant C, Tuariihionoa A, Bacou M, Souleyreau W, Sala M, Henriet E, Bikfalvi A, Saltel F, Auguste P. DDR1 and DDR2 physical interaction leads to signaling interconnection but with possible distinct functions. Cell Adh Migr. 2018;12(4):324–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Hammerman PS, Sos ML, Ramos AH, Xu C, Dutt A, Zhou W, Brace LE, Woods BA, Lin W, Zhang J. Mutations in the DDR2 kinase gene identify a novel therapeutic target in squamous cell lung cancer. Cancer Discov. 2011;1(1):78–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Tu HF, Chen MY, Lai JCY, Chen YL, Wong YW, Yang CC, Chen HY, Hsia SM, Shih YH, Shieh TM. Arecoline-regulated ataxia telangiectasia mutated expression level in oral cancer progression. Head Neck. 2019;41(8):2525–37.

    Article  PubMed  Google Scholar 

  51. GeneCards. Available from: https://www.genecards.org/cgi-bin/carddisp.pl?gene=ATM&keywords=ATM.

  52. Guérit E, Arts F, Dachy G, Boulouadnine B, Demoulin J-B. PDGF receptor mutations in human diseases. Cell Mol Life Sci: CMLS. 2021;78(8):3867–81.

    Article  PubMed  Google Scholar 

  53. Lin L-H, Lin J-S, Yang C-C, Cheng H-W, Chang K-W, Liu C-J. Overexpression of platelet-derived growth factor and its receptor are correlated with oral tumorigenesis and poor prognosis in oral squamous cell carcinoma. Int J Mol Sci. 2020;21(7):2360.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Shenoy S. CDH1 (E-Cadherin) mutation and gastric cancer: genetics, molecular mechanisms and guidelines for management. Cancer Manag Res. 2019;11:10477–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Pannone G, Santoro A, Feola A, Bufo P, Papagerakis P, Lo Muzio L, Staibano S, Ionna F, Longo F, Franco R, Aquino G, Contaldo M, De Maria S, Serpico R, De Rosa A, Rubini C, Papagerakis S, Giovane A, Tombolini V, Giordano A, Caraglia M, DiDomenico M. The role of E-cadherin down-regulation in oral cancer: CDH1 gene expression and epigenetic blockage. Curr Cancer Drug Targets. 2014;14(2):115–27.

    Article  CAS  PubMed  Google Scholar 

  56. Li G, Jiang W, Kang Y, Yu X, Zhang C, Feng Y. High expression of collagen 1A2 promotes the proliferation and metastasis of esophageal cancer cells. Ann Transl Med. 2020;8(24):1672.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Yu Y, Liu D, Liu Z, Li S, Ge Y, Sun W, Liu B. The inhibitory effects of COL1A2 on colorectal cancer cell proliferation, migration, and invasion. J Cancer. 2018;9(16):2953–62.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Tang M, Liu P, Wu X, Gong J, Weng J, Gao G, Liu Y, Gan L. COL3A1 and its related molecules as potential biomarkers in the development of Human Ewing’s Sarcoma. Biomed Res Int. 2021;2021:7453500.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Yuan L, Shu B, Chen L, Qian K, Wang Y, Qian G, Zhu Y, Cao X, Xie C, Xiao Y, Wang X. Overexpression of COL3A1 confers a poor prognosis in human bladder cancer identified by co-expression analysis. Oncotarget. 2017;8(41):70508–20.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Deng H, Huang X, Yuan L. Molecular genetics of the COL2A1-related disorders. Mutat Res/Rev Mutat Res. 2016;768:1–13.

    Article  CAS  PubMed  Google Scholar 

  61. Tarpey PS, Behjati S, Cooke SL, Van Loo P, Wedge DC, Pillay N, Marshall J, O’Meara S, Davies H, Nik-Zainal S, Beare D, Butler A, Gamble J, Hardy C, Hinton J, Jia MM, Jayakumar A, Jones D, Latimer C, Maddison M, Martin S, McLaren S, Menzies A, Mudie L, Raine K, Teague JW, Tubio JMC, Halai D, Tirabosco R, Amary F, Campbell PJ, Stratton MR, Flanagan AM, Futreal PA. Frequent mutation of the major cartilage collagen gene COL2A1 in chondrosarcoma. Nat Genet. 2013;45(8):923–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ganapathi MK, Jones WD, Sehouli J, Michener CM, Braicu IE, Norris EJ, Biscotti CV, Vaziri SAJ, Ganapathi RN. Expression profile of COL2A1 and the pseudogene SLC6A10P predicts tumor recurrence in high-grade serous ovarian cancer. Int J Cancer. 2016;138(3):679–88.

    Article  CAS  PubMed  Google Scholar 

  63. Tan Y, Chen Q, Xing Y, Zhang C, Pan S, An W, Xu H. High expression of COL5A2, a member of COL5 family, indicates the poor survival and facilitates cell migration in gastric cancer. Biosci Rep. 2021;41(4).

  64. Ding Y-L, Sun S-F, Zhao G-L. COL5A2 as a potential clinical biomarker for gastric cancer and renal metastasis. Medicine. 2021;100(7):e24561–e24561.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhou X-D, Qu Y-W, Wang L, Jia F-H, Chen P, Wang Y-P, Liu H-F. Identification of potential hub genes of gastric cancer. Medicine. 2022;101(41).

  66. Wang W, He Y, Zhao Q, Zhao X, Li Z. Identification of potential key genes in gastric cancer using bioinformatics analysis. Biomed Rep. 2020;12(4):178–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Mohammed A, Biegert G, Adamec J, Helikar T. Identification of potential tissue-specific cancer biomarkers and development of cancer versus normal genomic classifiers. Oncotarget. 2017;8(49):85692–715.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Wu L, Dong B, Zhang F, Li Y, Liu L. Prediction of the engendering mechanism and specific genes of primary melanoma by bioinformatics analysis. Dermatol Sin. 2016;34(1):14–9.

    Article  Google Scholar 

  69. Wu H, Liu Y, Duan H, Fan X, Wang Y, Song J, Han J, Yang M, Lu L, Nie G. Identification of differentially expressed circular RNAs in human nasopharyngeal carcinoma. Cancer Biomark. 2020;29(4):483–92.

    Article  CAS  PubMed  Google Scholar 

  70. Hermawan A, Putri H. Integrative bioinformatics analysis reveals potential target genes and TNFα Signaling Inhibition by Brazilin in Metastatic Breast Cancer Cells. Asian Pac J Cancer Prev. 2020;21(9):2751–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Jin X, Guan Y, Zhang Z, Wang H. Microarray data analysis on gene and miRNA expression to identify biomarkers in non-small cell lung cancer. BMC Cancer. 2020;20(1):329.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was part of an MSc thesis in Biostatistics. We would like to appreciate the Vice-chancellor of Education of the Hamadan University of Medical Science for technical support for their approval and support of this work.

Funding

This study was a part of the MSc thesis of the second author and it was supported (Grant No: 9807305722).

Author information

Authors and Affiliations

Authors

Contributions

LT conceived the research topic. LT and MGH and SA explored that idea, performed the statistical analysis, and drafted the manuscript. HM, AS, HKH participated in the interpretations and drafting of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Saeid Afshar.

Ethics declarations

Ethics approval and consent to participate

This study used a publicaly available data set. All methods were carried out in accordance with relevant guidelines and regulations, and the study was approved by the Ethics Committee of the Hamadan University of Medical Sciences (Ethical code: IR.UMSHA.REC.1398.618). The funding body had no role in the design of the study and collection as well as in writing the manuscript.

Consent for publication

Not applicable.

Competing interests

The authors have no conflicts of interest to declare for this study.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tapak, L., Ghasemi, M.K., Afshar, S. et al. Identification of gene profiles related to the development of oral cancer using a deep learning technique. BMC Med Genomics 16, 35 (2023). https://doi.org/10.1186/s12920-023-01462-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12920-023-01462-6

Keywords