Abstract Hepatocellular carcinoma (HCC) is an aggressive and chemoresistant cancer type. The development of novel therapeutic strategies is still urgently needed. Personalized or precision medicine is a new trend in cancer therapy, which treats cancer patients with specific genetic alterations. In this study, a gene signature was identified from the transcriptome of HCC patients, which was correlated with the patients’ poorer prognoses. This gene signature is functionally related to mitotic cell cycle regulation, and its higher or lower expression is linked to the mutation in tumor protein p53 (TP53) or catenin beta 1 (CTNNB1), respectively. Gene–drug association analysis indicated that the taxanes, such as the clinically approved anticancer drug paclitaxel, are potential drugs targeting this mitotic gene signature. Accordingly, HCC cell lines harboring mutant TP53 or wild-type CTNNB1 genes are more sensitive to paclitaxel treatment. Therefore, our results imply that HCC patients with mutant TP53 or wild-type CTNNB1 genes may benefit from the paclitaxel therapy. Keywords: bioinformatics, hepatocellular carcinoma, precision medicine, taxanes 1. Introduction Primary liver cancer is still the sixth most common cancer type and the third leading cause of cancer-related death in the world [[34]1]. Most (75–85%) of primary liver cancer cases involve hepatocellular carcinoma (HCC), whose major risk factors include chronic hepatitis B or C virus (HBV or HCV) infection, aflatoxin contamination in foods, excess body weight, heavy alcohol intake, smoking, and type 2 diabetes [[35]1]. HCC development involves a complex, multi-step histological process of normal hepatocyte malignant transformation involving various genetic and epigenetic alterations [[36]2]. The most frequent genetic alterations include mutations in telomerase reverse transcriptase (TERT) promoter, tumor protein p53 (TP53), and catenin beta 1 (CTNNB1) genes, as well as copy number variations and aberrations in DNA methylation [[37]3]. TERT promoter mutations occur in dysplastic nodules and early HCC, and this gene is viewed as a gatekeeper for malignant transformation. TP53 and CTNNB1 mutations function as drivers during HCC development. Additional molecular alterations include focal DNA amplification (for example, vascular endothelial growth factor A/VEGFA, MYC proto-oncogene, bHLH transcription factor/MYC) and deletions (for example, cyclin-dependent kinase inhibitor 2A/CDKN2A, axin 1/AXIN1), and DNA methylation of promoter regions [[38]3]. The current options for HCC management include surgical resection, liver transplantation, percutaneous local ablations (such as ethanol injection and radiofrequency thermal ablation), transarterial chemoembolization, transarterial radioembolization, and systemic pharmacological therapies [[39]4,[40]5]. The main curative treatments for HCC are surgical resection and liver transplantation, which are, however, only suitable for 15% to 25% of patients [[41]6]. Furthermore, HCC is a chemoresistant and extremely refractory tumor type, and no reliable and effective treatments are available for those with advanced or metastatic disease [[42]6]. Molecular targeted agents and immunotherapy have been regarded as treatment options in recent years. Although several multi-kinase inhibitors, such as sorafenib, regorafenib, lenvatinib, and cabozantinib, have been approved for treating advanced HCC [[43]7,[44]8,[45]9,[46]10], they only provide a short increase in median overall survival [[47]7,[48]8,[49]10,[50]11,[51]12]. Immune checkpoint inhibitors, such as human anti-PD-1 monoclonal antibodies (nivolumab and pembrolizumab) and human anti-CTLA4 monoclonal antibody (ipilimumab), were approved for advanced HCC from 2017 to 2020, which greatly extend the patients’ overall survival [[52]13,[53]14,[54]15]. Personalized or precision medicine has become a new trend in cancer treatment, which helps doctors select treatment for patients based on their genetic alterations [[55]16]. Because HCC is a highly heterogeneous disease, grouping HCC patients into relatively homogeneous molecular subtypes may offer a significant clinical benefit through precision treatment [[56]17]. HCC is usually classified based on tumor burden [[57]18]. Recent advances in multi-omics technologies provide an opportunity for developing personalized treatment against HCC. For example, next-generation sequencing analyses of HCC identified new mutational signatures and defined new tumor subtypes that may benefit from targeted treatments in the future [[58]19,[59]20]. In this study, an integrated bioinformatics analysis determined that a prognostic mitotic gene signature is associated with TP53/CTNNB1 mutation statuses in HCC. This gene signature could be targeted by paclitaxel, a clinically approved anticancer drug. Our results support that HCC patients with mutant TP53 or wild-type CTNNB1 genes may benefit from the paclitaxel anticancer therapy. 2. Materials and Methods 2.1. Cancer Genomics Analysis Four microarray data sets from HCC patients, including [60]GSE14520 [[61]21], [62]GSE45267 [[63]22], [64]GSE50579 [[65]23], and [66]GSE62232 [[67]19], were obtained from the public Gene Expression Omnibus (GEO) depository database at the National Center for Biotechnology Information (NCBI). The differentially expressed genes (DEGs) between HCC tumor and adjacent normal tissues were obtained using the GEO2R online tool [[68]24], and the criteria used to define DEGs were as follows: an adjusted p-value < 0.01 and |Log[2] fold-change| > 1. The full DEG list is shown in [69]File S1. The overlapped genes among four microarray data sets were visualized by a heat map generated using MORPHEUS software ([70]https://software.broadinstitute.org/morpheus/; accessed on 21 January 2021). The prognostic values of these overlapped genes in HCC patients were further explored by GEPIA2 ([71]http://gepia2.cancer-pku.cn/; accessed on 21 January 2021) [[72]25] and/or cBioPortal ([73]https://www.cbioportal.org/; accessed on 21 January 2021) [[74]26,[75]27], using The Cancer Genome Atlas (TCGA) hepatocellular liver carcinoma (LIHC) data set. 2.2. Pathway Enrichment Analysis For pathway enrichment in Gene Ontology (GO) biological process, Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome, selected genes were analyzed by STRING ([76]https://string-db.org/; accessed on 21 January 2021), a database of known and predicted protein–protein interactions based on computational prediction, knowledge transfer between organisms, and interactions aggregated from other primary databases [[77]28]. The following parameters were used: organism = Homo sapiens; network type = full network; network edges = evidence; active interaction sources = experiments and databases; minimum required interaction score = 0.4; max number of interactors to show = queried proteins only. 2.3. Gene–Drug Association Analysis Gene–drug association was analyzed using GLAD4U ([78]http://dlad4u.zhang-lab.org/; accessed on 26 January 2021) [[79]29] via the WebGestalt website ([80]http://www.webgestalt.org/; accessed on 26 January 2021) [[81]30]. GLAD4U is a gene retrieval and prioritization tool based on existing biomedical literature [[82]29]. WebGestalt is a functional enrichment analysis web tool that integrates various primary databases, including GLAD4U [[83]30]. The gene–drug interaction network was further constructed using STITCH ([84]http://stitch.embl.de/; accessed on 26 January 2021), a database of known and predicted interactions between chemicals and proteins based on computational prediction, knowledge transfer between organisms, and interactions aggregated from other primary databases [[85]31]. The following parameters were used: organism = Homo sapiens; network edges = evidence; active interaction sources = experiments and databases; minimum required interaction score = 0.4; max number of interactors to show = queried proteins only. 2.4. Cancer Cell Drug Sensitivity Analysis The drug sensitivity data, gene mutation status, and gene expression levels in HCC cell lines ([86]Table S1) were downloaded from CellMinerCDB ([87]https://discover.nci.nih.gov/cellminercdb/; accessed on 25 July 2021) [[88]32] using the data in the Cancer Therapeutics Response Portal (CTRP; [89]https://portals.broadinstitute.org/ctrp.v2.1/; accessed on 25 July 2021) [[90]33,[91]34,[92]35]. The CTRP database links cancer cells’ genetic features to drug sensitivity [[93]33,[94]34,[95]35], and CellMinerCDB is an interactive web-based tool allowing integration and analysis of genetic and pharmacological data in cancer cell lines across various data sets, including the CTRP [[96]32]. 3. Results 3.1. Identification of a Prognostic Mitotic Gene Signature in Hepatocellular Carcinoma To identify a common gene signature in HCC patients, four microarray data sets ([97]Table 1) were employed, and the common DEGs and their fold-change values are visualized in [98]Figure 1 (left). To characterize the most essential genes in HCC patients, the prognostic roles of these DEGs in patients’ overall survival were analyzed using the TCGA-LIHC data set. As shown in [99]Figure 1 (right), 30 upregulated and 2 downregulated genes were significantly linked to the patients’ overall survival. The interactions of these 32 genes were constructed by STRING [[100]28]. Pathway enrichment results found that most of the upregulated genes are related to the cell cycle procession, especially mitosis ([101]Figure 2). For example, mitosis is initiated by activating cyclin-dependent kinase 1 (CDK1) with its binding partner, cyclin B1 (CCNB1). Aurora kinase A (AURKA) and NIMA (never in mitosis gene A)-related kinase 2 (NEK2) are also mitotic kinases. Once activated, these mitotic kinases simultaneously or sequentially phosphorylate more than 1000 mitotic substrates (such as cell division cycle 20/CDC20 and pituitary tumor transforming gene 1/PTTG1) to regulate mitotic progression [[102]36]. These results indicate that the commonly upregulated genes contributing to poor prognoses of HCC patients are correlated with the aberrant regulation of mitosis. Table 1. Microarray data sets for hepatocellular carcinoma patients. Access Number Platform Normal Tumor References