Open Access

Gene module analysis of juvenile myelomonocytic leukemia and screening of anticancer drugs

  • Authors:
    • Wencheng Zhao
    • Lin Wang
    • Yongbin Yu
  • View Affiliations

  • Published online on: September 18, 2018     https://doi.org/10.3892/or.2018.6709
  • Pages: 3155-3170
  • Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Juvenile myelomonocytic leukemia (JMML) is a rare but severe primary hemopoietic system tumor of childhood, most frequent in children 4 years and younger. There are currently no specific anticancer therapies targeting JMML, and the underlying gene expression changes have not been revealed. To define molecular targets and possible biomarkers for early diagnosis, optimal treatment, and prognosis, we conducted microarray data analysis using the Gene Expression Omnibus, and constructed protein‑protein interaction networks of all differentially expressed genes. Modular bioinformatics analysis revealed four core functional modules for JMML. We analyzed the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway functions associated with these modules. Using the CMap database, nine potential anticancer drugs were identified that modulate expression levels of many JMML‑associated genes. In addition, we identified possible miRNAs and transcription factors regulating these differentially expressed genes. This study defines a new research strategy for developing JMML‑targeted chemotherapies.

Introduction

Juvenile myelomonocytic leukemia (JMML) is an infrequent but aggressive hematological system tumor of infancy and childhood with poor prognosis. Signs and symptoms include fever, thrombocytopenia, hemorrhage, lymphadenopathy, high fetal hemoglobin levels and progressive hematologic anemia, hepatosplenomegaly, and clonal proliferation of myelomonocytic cells (13). The most effective treatment currently available is allogeneic hematopoietic stem cell transplantation (HSCT). However, the overall survival at 5 years after transplant is only 52–63% due to treatment-related toxicity and frequent disease relapse (37). In addition, some patients lack a suitable human leukocyte antigen-matched donor, thus novel chemotherapeutic regimens are needed to further improve outcome.

Malignant tumors are a major cause of mortality in children, and traditional cancer treatments such as chemotherapy, radiotherapy and surgery carry severe side effects or do not markedly improve prognosis. As an aggressive myeloproliferative neoplasm, response to conventional chemotherapy is weak, thus most JMML patients require early allotransplantation (6,7). Alternatively, gene module analysis of JMML may facilitate the screening of anticancer drugs targeting specific anomalies in gene expression. In the present study, we performed gene module analysis to identify gene expression signatures of JMML and to illuminate the functions of these altered genes and protein networks in disease pathogenesis.

While there are voluminous studies on anticancer drug pharmacokinetics and efficacy against solid tumors, there have been few studies focusing on JMML. Drugs that target JMML-associated genes can be an effective anticancer approach, either alone or in combination with traditional modalities. Modular analysis of bioinformatic data is emerging as a valuable strategy for systematic and comprehensive analysis of regulatory signaling pathways relevant to disease etiology and drug treatment effects (8,9). In the present study, we used modular analysis bioinformatics methods to search for differentially expressed genes and their key functional group(s) and networks in JMML.

Genes differentially expressed in leukemic cells compared to normal cells were classified into upregulated and downregulated groups. These differentially expressed genes were then compared to differentially expressed genes in cells treated with small molecules from the CMap database by converting them into probe setters based on the HG-U133A platform. Genes were then assigned enrichment values between-1 and 1 according to whether specific small molecules stimulated the expression state in leukemia cells (closer to 1) or normal cells (closer to −1). Based on this analysis, we identified drugs that regulate key nodes of JMML-associated regulatory networks. The large number of genes and drugs identified provide many possible targets and effective chemotherapies for JMML.

Materials and methods

Screening for differentially expressed genes in JMML

We downloaded JMML microarray data from the GEO platform (https://www.ncbi.nlm.nih.gov/geo/) [data code GSE71449 (human)] (10).

The criteria for data selection were gene chip expression profiling data accrued over the last three years (since 2014) with clear disease and normal control groups in the data set. The disease queried was ‘Juvenile myelomonocytic leukemia’. The largest number of GSE71449 samples were obtained by such a standard. While our analysis is indeed limited to all disease-related data sets in the database, further analysis of other data sets, including miRNA chips, is already under way as no single data set represents all genes associated with a disease. The mechanisms of any disease are only revealed by direct experimental study, but these gene chip results may identify genes with greater probability of involvement. The single-chip DEG analysis can only reveal that in the current data set, these differentially expressed genes (DEGs) are most likely to be related to disease.

Array Express was retrieved, and the results retrieved with the same search criteria coincided with those in GEO. There was no larger sample size under the same screening criteria. GEO data sets are included in the EBI database, thus there is no mention of it.

The database provides tissue sample results from 44 JMML patients and seven healthy donors obtained using the Agilent-041648 CMGG Human V1.1 60k [Probe Name Version] microarray platform. We used the R software bagaffy (11) (Version1.50.0, http://www.bioconductor.org/packages/release/bioc/html/affy.html) to read the downloaded microarray matrix data and the robust multi-array average method (12,13) to perform standardized data preprocessing (including Background correction, Normalization, and Expression calculation). We performed annotation for probes in the platform annotation file and deleted probes that did not match the gene symbols used. When the same gene was reflected by different probes, we adopted the average as the final expression value.

For the two data groups (JMML and controls), we screened for differentially expressed genes using the R bag Limma microarray analysis package (14) and adopted the BH-corrected T test to identify genes significantly over- or under expressed. For every differentially expressed gene, we assumed a threshold P<0.05 and log|FC| >0.58. Differentially expressed genes were selected based on a threshold |logFC| >0.58, corresponding to a fold change >1.5 or ≤1.5 (15). We then used the common enrichment analysis tool DAVID (16) (version 6.8) and subjected genes with enrichment ≥2 and significance threshold of hypergeometric test P<0.05 to Gene Ontology (GO) (17) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) (18) pathway analyses.

For observing the function of differentially expressed genes intuitively, we used the ClueGO (19) plug-in (version 2.2.6, http://apps.cytoscape.org/apps/ClueGO) of Cytoscape software (20) and constructed separate GO BP and KEGG pathway cross-linked enrichment graphs for upregulated and downregulated genes using P<0.05 as the significantly enriched threshold.

Building and analyzing modules of differentially expressed genes

We used the STRING (21) database bank to predict possible protein-protein interaction (PPI) networks for all differentially expressed proteins and genes with parameter PPI score 0.4 (medium confidence). Networks with average interaction strengths at protein nodes were constructed using Cytoscape.

Interaction networks may contain nodes in which the interactions are closed among the constituent genes. These nodes thus represent distinct biological processes. There are many methods to define nodes by PPI cluster analysis but MCODE (22) was chosen because it is the most common. We applied MCODE to calculate the scores of every node, with higher scores indicating a greater degree of separation from other nodes and stronger association with specific processes. We choose nodes with score ≥5 and node number ≥5 for subsequent GO and KEGG pathway analyses.

Screening for drugs that regulate functional modules

The CMap database (23,24) stores the genome-wide expression profiles of human cells treated with various active small molecules. In total, CMap contains data from 6,100 small molecule interference experiments (with normal control groups) using 1,309 small molecules, for a total of 7,056 different expression profiles.

We analyzed gene expression differences between normal cells and leukemia cells and then compared the responses of differentially expressed genes to identify those with similar or opposite effects (upregulation vs. downregulation) on normal cells. We divided the genes differentially expressed between normal and leukemia cells into upregulated and downregulated subgroups. Then we transformed the HG-U133A platform probe set results and compared them to the set of differentially expressed genes under small molecule treatment from the CMap database to obtain enrichment values. In this case, enrichment values range from-1 and 1, with those closer to 1 influenced to a greater extent by small molecules in normal cells and those with values closer to −1 influenced to a greater extent by small molecules in leukemia cells.

For key modular gene screening, we constructed connectivity maps and used the gene expression differences in human cells treated with small molecules to identify drugs affecting the expression levels of genes associated with the disease.

Identification of possible miRNAs regulating target genes

We identified potential miRNA targeting genes within PPIs using Webgestalt (http://www.webgestalt.org/) (25) tools and overrepresentation enrichment analysis methods with a threshold P<0.05 by hypergeometric tests and BH correction for enriched gene number and count ≥5.

Identification of possible transcription factors regulating target genes

Based on the transcription factor-regulated network data in the ITFP database and TRANSFAC bank, we searched for transcription factors regulating differentially expressed genes and further screened the differentially expressed target genes regulated by these transcription factors for network integration. We performed network integration for the obtained TF-Target, miRNA-Target, and PPI network and constructed integration networks using Cytoscape.

Statistical analysis

t-tests were used to screen for differentially expressed genes and to evaluate the enrichment obtained by GO_BP- and KEGG-based methods. A P<0.01 was considered statistically significant. With respect to each herbal component, Fisher's exact test was used to evaluate the genes in modules. Based on the above data, we set P<0.01 for each component.

Results

Screening for differentially expressed genes in JMML

We downloaded the JMML expression data from the GEO database, and identified 700 upregulated and 737 downregulated genes at threshold levels of P<0.05 and log|FC| >0.58. From these genes, we constructed heat maps (Fig. 1). We then performed GO and KEGG pathway enrichment analyses for upregulated and downregulated mRNA muster, and according to the screening threshold value obtained GO terms and KEGG pathways including these differentially expressed genes. Fig. 2A shows the top five GO Biological Process (BP) terms and KEGG pathways including upregulated mRNAs and Fig. 2B shows these same results for downregulated mRNAs.

We used clueGO to perform GO and KEGG pathway analysis for all upregulated and downregulated differentially expressed genes (Fig. 3A and B). ClueGO provides kappa coefficients that can be used to divide the pathways into functional groups. In the figure, different colors indicate pathway enrichment results and correlations between two pathways are reflected by a connecting line. The sizes of nodes reflect the P-value. Fusion of enrichment results was performed when different terms were enriched for the same gene. In the figure, GO terms are indicated by different colors. Thus, one color indicates a functional group. The size of the term nodes depends on the threshold P-value, increasing as P gets smaller.

Key genes and PPI networks constructed from differentially expressed genes

Combined with the PPI database, we constructed the network figures for differentially expressed genes (Fig. 4). The PPI network of differentially expressed genes contained 908 nodes and 5,053 related pairs. The relationships among the nodes are closed. Table I lists the top 10 highest degree nodes obtained from the PPI network and Fig. 4 shows the PPI network constructed from the differentially expressed genes, with pink nodes indicating upregulated and blue nodes the downregulated genes.

Table I.

The top 10 highest degree nodes obtained from the PPI network.

Table I.

The top 10 highest degree nodes obtained from the PPI network.

GeneDegreeBetweennessCloseness
MAPK1283.0142874.90.54246414
CD4218.093239.10.49266702
OAS3212.0110066.460.48373333
CD79A173.059164.6330.47862798
NFKBIA157.037323.4530.48424986
BCL6154.042434.6840.4748691
EGR2138.033878.510.4512438
PLEK134.053153.110.45509282
TRIB3115.024556.3030.44591936
RASL11A112.016269.2450.44923228

[i] PPI, protein-protein interaction.

Modular analysis

We obtained a total of 10 modules from the PPI network, of which four had scores >3 (Fig. 5). We also performed functional enrichment analysis for the genes of modules. Fig. 6 shows the top five GO BP terms and KEGG pathways in module 1, the four KEGG pathways and top five GO BP terms in module 2, the four GO BP terms in module 3 (no KEGG pathways were enriched), and the top five GO BP terms and KEGG pathways in module 4.

For module 1, pathway analysis indicated that differentially expressed genes were most strongly (lowest P-value) related to Regulation of cell proliferation. For module 2, differentially expressed genes were most strongly related to Pathways in cancer. In module 3, the strongest relationship was with Transcription, DNA-templated, and in module 4 the strongest relationship was with Immune response.

Small drug molecules related to diseases

We performed small molecule drug analysis for the 4 modules using the CMap database and threshold P<0.05. The regulatory effects on each module are shown in Fig. 7.

Statistical relationships between modular genes and small drugs

We used local perl script and performed Pubmed searches for the analyzed modular genes and the corresponding small molecules, and constructed the network figures shown in Fig. 8. The P-values were considered in descending order, and the drugs with the 10 smallest P-values were used for subsequent analyses. A total of 40 drugs were identified as possible regulators of the 4 modules. To obtain novel anticancer drugs, we conducted a literature review of the cancer-related research on the 24 drugs with greatest effects on gene expression. In cancer treatment, the number of publications reflects the extent of research on a given drug's potential efficacy. Multiple drugs have been researched extensively and already certified to be effective anticancer drugs, such as etoposide and phosphonothreonine (the number of relevant publications was >100). Others, however, have not been fully studied for their anticancer efficacies, such as disulfiram, ursolic acid, miconazole, thioridazine, loperamide, monastrol (the number of relevant publications was ≤10), and others, such as nadolol, tetracaine and levomepromazine have never been studied with respect to cancer. Publication numbers for drug are shown in Fig. 9.

Identification of candidate miRNAs regulating differentially expressed genes

We identified candidate miRNAs that may regulate the differentially expressed genes using a screening threshold of P<0.05. For upregulated genes, we identified 10 candidate miRNAs (Table IIA) and for the downregulated genes four candidate miRNAs (Table IIB).

Table II.

Identification of candidate miRNAs regulating differentially expressed genes (DEGs).

Table II.

Identification of candidate miRNAs regulating differentially expressed genes (DEGs).

A, miRNAs for upregulated DEGs

miRNAP-value
AGTCAGC, MIR-3450.002846737
CAGGTCC, MIR-4920.003876902
ACCAATC, MIR-5090.011602863
CCAGGTT, MIR-4900.017555181
TGCACTG, MIR-148A, MIR-152, MIR-148B0.023681008
TGAGATT, MIR-2160.030489785
TCCAGAT, MIR-516-5P0.033806763
AGTCTTA, MIR-4990.034998955
GTGTCAA, MIR-5140.037442107
AAAGGGA, MIR-204, MIR-2110.040666691

B, miRNAs for downregulated DEGs

miRNAP-value

GGCACTT, MIR-519E0.002921036
TCCGTCC, MIR-1840.037734308
GTAAGAT, MIR-200A0.040359704
AGCATTA, MIR-1550.04249545
Identification of candidate transcription factors regulating differentially expressed genes

For the upregulated differentially expressed genes, we identified 22 differentially expressed transcription factors (TFs). For the downregulated differentially expressed genes, we identified 19 differentially expressed TFs. The results are summarized in Table III.

Table III.

Identification of candidate transcription factors (TFs) regulating differentially expressed genes (DEGs).

Table III.

Identification of candidate transcription factors (TFs) regulating differentially expressed genes (DEGs).

TFs for upregulated DEGsTFs for downregulated DEGs
ABTB2, AHCTF1, BATF, CORO7, ERG, FGD5, FOXN2, IFIH1, IFIT1, IFIT2, IFIT3, ING1, INPP5A, LASP1, PARP14, PLEK, PTBP2, SAMD9, STAT1, TANK, TTC7B, WDR1CCNE1, CDC20, CKAP5, FAM129B, KRT7, LGR4, MKL2, NFIA, REXO1, SMARCA4, SPEN, TLE2, TRIM10, TRIM27, TTC16, ZBTB4, ZNF497, ZNF750, ZNF771

Discussion

Currently the only curative treatment option for JMML is hematopoietic stem cell transplantation (HSCT). However, disease recurrence remains a major cause of treatment failure (7). Clinical symptoms are caused by hematopoietic insufficiency and excessive proliferation of leukemic monocytes and granulocytes, leading to hepatosplenomegaly, lymphadenopathy, skin rash and respiratory failure (3,7,26). A serious obstacle to the research of JMML is the lack of suitable experimental models, impeding the development and pre-clinical evaluation of novel therapeutic approaches. Primary JMML leukemia cells cannot be maintained in culture as they differentiate and become apoptotic (27), while an immortalized cell line derived from JMML cells has not yet been successfully established (28). The generation of induced pluripotent stem cell lines originating from JMML cells was reported, but conceptually such systems are limited by their artificial nature and the risk of further transformation during reprogramming (29). Therefore, it is important to identify tumor-promoting processes, such as dysregulation of specific genes and networks, and develop targeted therapies.

In the present study, we combined the genome-wide gene chip information already available from JMML tissues and used modular analysis bioinformatics procedures to identify molecular targets and small molecule compounds that affect their expression. For this purpose, modular analysis is a valuable and well established bioinformatic method that is useful for analysis of both large-scale protein networks and single proteins and genes. Moreover, this method has been used to study the development and treatment of multiple diseases. After the enrichment analysis, we found that genes related to the hematopoietic cell lineage and to regulation of immune responses were significantly downregulated, while chemokine signaling pathway and inflammatory response genes were significantly upregulated compared to healthy tissue.

Hematopoietic progenitors produce a myriad of diverse lineages, including progenitors with lymphoid, myeloid, and erythroid potential, prior to hematopoietic stem cells. Previous pathway analyses revealed that genes related to hematopoietic cell lineages were significantly downregulated in JMML (3034), and we further found that genes related to regulation of immune responses were also significantly downregulated. Anticancer treatment leads to dense immune infiltrate, including many CD8+ and CD4+ cells, concomitant with tumor regression, at both treated and untreated lesions, consistent with generation of a tumor-specific systemic immune response (35). Several studies have indicated the synergistic potential of various immunomodulatory agents (3639). Therapies that enhance or impede immune responses are essential but optimal timing and the administration route are important detemising results, such as increased survival and immune responses, leading to complete tumor regression in some cases (4042).

Rminants of efficacy. Preclinical studies combining these agents have shown proThe PI3K/Akt and MAPK signaling pathways have been shown to mediate chemokine-induced migration of multiple cell types. Modular analysis suggested that the Chemokine signaling pathway (module 1) was strongly related to JMML morbidity. In addition to regulation of cell proliferation, differentiation, invasion, and inflammation, chemokines are widely involved in the regulation of cancer. Analysis of module 1 revealed that regulation of cell proliferation was significantly increased and strongly related to JMML occurrence. Further, analysis of all 4 functional modules revealed complex interactions among a number of genes involved in Chemokine signaling pathway and Regulation of cell proliferation. Chemokines are small proteins expressed in response to injury or infection and during normal immune surveillance. Chemokines are involved in leukocyte trafficking and regulate tumor metastasis, proliferation, differentiation and angiogenesis (4346).

Module 2 contained genes primarily involved in Pathways in cancer, Transcriptional dysregulation in cancer, and Signal transduction, suggesting that module 2 contains clusters of key tumor-promoting genes. Accumulation of driver somatic alterations in genes modifies critical cellular processes leading to cancer (47,48). In recent years, the catalog of driver genes known to take part in the development of malignancies has expanded due to whole-exome and whole-genome analyses of large tumor sets by large international consortia (49,50). In addition to upregulation of oncogenes, downregulation of anticancer proteins or dysregulation of subcellular localization may also contribute to JMML.

Cancer cells scatter from tumors in hoards and build new tumors in distant tissues and organs (51,52). In over 90% of fatal cancers, metastasis is the cause of death. Tumors contain a heterogeneous population of cells in an organized hierarchy akin to normal tissue. Tumor progression, invasiveness, and self-renewal are attributes of smaller subfractions of cancer cells. Tumor initiation by disseminated cancer cells relies on their ability to self-renew and initiate metastatic tumors.

Genes in module 3 were primarily involved in Response to hypoxia, Negative regulation of transcription from RNA polymerase II promoter, Regulation of transcription, DNA-templated, and Transcription, DNA-templated. We found that among these pathways, transcription, DNA-templated was most strongly related to JMML occurrence. The identification and detection of specific nucleic acids (either DNA or RNA) is an enabling technology for forensic analysis (53), recognition of genetic mutations (54) and pathogen identification (55). For many methods, quantification of nucleic acids is important for identifying a given DNA sequence. For example, in many forms of cancer (56), oncogene copy number increases and deletion of tumor-suppressor genes can be found. In addition to measuring changes in gene copy number, quantitative detection of circulating DNAs can be a useful diagnostic tool for identifying cancer (57).

The genes in module 4 are closely associated with extracellular matrix organization and immune responses. Much effort has been devoted to determining how cellular components of the tumor promote cancer development and initiate formation of a niche conducive to growth (58). Alternatively, the importance of non-cellular components of the niche during cancer progression is well documented, particularly extracellular matrix (ECM) organization (5962). In addition to a stable structure with supportive functions in maintaining tissue morphology, the ECM is a surprisingly dynamic and versatile part of the cell milieu influencing fundamental aspects of cell biology (63). For major developmental processes, the ECM directly or indirectly regulates almost all cellular behaviors (6467). Indeed, in diseases such as cancer, abnormal ECM dynamics may be a strong determinant of clinical outcome (68). How disruption of ECM dynamics contributes to tumor development is a challenging issue in cancer biology.

The local immune response has also emerged as an important element in the multistep process of cancer development (69). Observations that some tumors arise from chronic inflammation sites have led to speculation of a strong connection between tumor onset and inflammatory pathways. In addition, some tumors are infiltrated by both the innate and adaptive arms of the immune system, thus many different immune cells are present within the tumor microenvironment (70). Elements of both the innate and adaptive immune systems have been reported to act both as pro- or anti-tumorigenic factors depending on the relative balance. The intercellular communication between infiltrating immune cells and cancer cells modulates this immune response so as to positively influence tumor development (71).

We found the module 2 was most strongly related to JMML occurrence. There are likely synergistic interactions among module 2 genes.

Many investigations have attempted to screen for well-tolerated and affordable anti-neoplastic medications, and a myriad of drugs with cytotoxicity against human cancer cell lines have been described (72,73). In our study, we also screened for drugs with curative potential against JMML through modulation of associated gene pathways. In 1999, Mayer et al (74) first identified monastrol, a cell-permeable small molecule with antimitotic activity but without neuronal cytotoxicity. Through inhibition of kinesin Eg5, monastrol can induce the mono-astral conformation of microtubules (75,76). Several subsequent studies have clarified the anti-mitotic mechanisms of monastrol (7779), but few studies have investigated its anticancer activity (8082).

In both canine and human cancer patients, the peripherally acting µ-opiate receptor agonist loperamide hydrochloride is recommended as a treatment for chemotherapy-related diarrhea (83,84). Loperamide was shown to dose-dependently induce apoptosis and suppress the proliferation of human liver, lung, bone, and breast cancer cell lines (85). In human cancer cell lines, the mechanism underlying apoptosis induction has not been fully elucidated, although the caspase 3 pathway has been implicated (85) and loperamide will exert anticancer properties at clinically relevant doses. In the clinic, loperamide would be an attractive drug for JMML because of its minimal side effects and low price. Our bioinformatic analysis revealed a potential role for loperamide in JMML treatment. Further studies are needed to establish mechanisms of antitumor activity.

Originally, thioridazine (TDZ) was used as a therapy for psychotic disease (86,87). In addition, it has been used to treat drug-resistant microorganisms (88,89). Recently TDZ was reported to have potent effects on various types of cancer cells, including anti-angiogenesis and apoptosis promotion of breast and ovarian cancers cells (90,91). In addition, TDZ induced cytotoxicity of cervical (92), prostate (93), gastric (94), and pancreatic cancer cells (95). TDZ also has selectivity in leukemia as a dopamine receptor inhibitor (96,97).

A few publications have suggested that miconazole has anticancer effects (98). Miconazole is a common treatment for superficial fungal infection and a prominent systemic antifungal agent. In different human neoplastic cell lines, Wu et al demonstrated that miconazole could induce cell cycle arrest. This growth arrest was dose-dependent and related to the p53 signaling pathway (99). Anticancer effects of miconazole were reported on 4T1 (breast cancer) and 5637 (bladder cancer) cell lines.

Ursolic acid (UA) was found to have a biphasic response against three breast cancer cell lines (100). Another study (101) reported that disulfiram suppressed tumor growth by killing cancer cells and was even effective in combination with DHA. Indeed, UA could act as a substitute for clioquinol.

Compared to treatments targeting individual genes, treatment with agents affecting larger gene groups may have better efficacy as anticancer therapy. The aim of the present research was to identify possible molecular targets for cancer treatment and potential anticancer drugs. We searched for drugs that regulate the essential functional modules of JMML using the DrugBank Small Molecule database, and found several drugs already shown to have anticancer effects, including nadolol, disulfram, ursolic acid, micronazole, thioridazine, loperamide, monastrol, tetracaine, and levomepromazine. In contrast, other drugs identified have never been examined for anticancer efficacy, such as tetracaine, levomepromazine and nadolol. Nonetheless, effects on differentially expressed genes in JMML suggest therapeutic potential.

The nine drugs identified are potential therapies for JMML. Many widely used and studied drugs have never been examined for effects against cancer, such as tetracaine, levomepromazine and nadolol. According to our enrichment analyses, these agents hold potential for JMML treatment. We searched the Pubmed database and found several RCTs on these drugs, but no published clinical RCTs supporting anticancer effects on JMML. In the future, we plan to perform RCTs on these nine drugs examining possible therapeutic efficacy against JMML.

When designing therapeutic regimens for JMML, possible adverse reactions must be considered. It is thus noteworthy that the drugs identified here are in clinical use with well described safety profiles. When tumor cell proliferation reaches the highest activity, drugs interfering with the cell cycle or targeting proliferation pathways are significantly more potent.

MicroRNAs are endogenous 20–25 nucleotide non-coding RNAs found in eukaryotes that regulate gene expression at the level of translation. Mature miRNAs are produced by sequential cleavage of longer primary transcripts. Then the RNA-induced silencing complex RISC induced by RNA is assembled to recognize target mRNAs by base complementary pairing, and the target mRNA is degraded or suppressed according to the degree of complementarity. Recent studies have shown that the silencing complex degrades the target mRNA or inhibits the translation of the target mRNA. Through gene silencing, miRNAs regulate diverse processes including growth, virus defense, hematopoiesis, organ formation, cell propagation, apoptosis, and fat metabolism, among others. Transcription factors (TFs) are proteins with special structures that regulate gene expression at the level of the genome. The transcription initiation of eukaryotes is complex and requires the assistance of multiple protein cofactors. Transcription factors form a transcription initiation complex with RNA polymerase II. Both miRNAs and TFs regulate genes, thus differentially expressed genes (DEGs) are predicted by miRNAs and TFs, which further extends the number of potential targets. Like differentially expressed genes, the prediction of miRNAs and TFs involved in differential expression also provides clues to disease mechanisms.

In conclusion, our study used multiple bioinformatic methods to identify 4 gene modules associated with JMML. In addition, we identified 40 drugs based on the CMap database that can alter expression and function of the 4 modules. We performed Fisher's exact test to precisely screen for drugs with the strongest effects on module regulation. Through our study, we identified nine drugs that have considerable potential as new anticancer drugs. Moreover, the study provides a new research template for future research on JMML-targeted anticancer treatments through detailed analyses of core functions. In the future, we will conduct follow-up studies to verify the anticancer effects of the selected drugs.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Research Project of Heilongjiang Health and Family Planning Commission (project no. 2016-038).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

WZ and YY conceived and designed the study. WZ, LW and YY performed the experiments. WZ and LW wrote the manuscript. WZ, LW and YY reviewed and edited the manuscript and were also involved in the conception of the study. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chan RJ, Cooper T, Kratz CP, Weiss B and Loh ML: Juvenile myelomonocytic leukemia: A report from the 2nd International JMML Symposium. Leuk Res. 33:355–362. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Niemeyer CM and Kratz CP: Paediatric myelodysplastic syndromes and juvenile myelomonocytic leukaemia: Molecular classification and treatment options. Br J Haematol. 140:610–624. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Niemeyer CM, Arico M, Basso G, Biondi A, Rajnoldi Cantu A, Creutzig U, Haas O, Harbott J, Hasle H, Kerndrup G, et al: Chronic myelomonocytic leukemia in childhood: A retrospective analysis of 110 cases. European working group on myelodysplastic syndromes in childhood (EWOG-MDS). Blood. 89:3534–3543. 1997.PubMed/NCBI

4 

Bergstraesser E, Hasle H, Rogge T, Fischer A, Zimmermann M, Noellke P and Niemeyer CM: Non-hematopoietic stem cell transplantation treatment of juvenile myelomonocytic leukemia: A retrospective analysis and definition of response criteria. Pediatr Blood Cancer. 49:629–633. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Dvorak CC and Loh ML: Juvenile myelomonocytic leukemia: Molecular pathogenesis informs current approaches to therapy and hematopoietic cell transplantation. Front Pediatr. 2:252014. View Article : Google Scholar : PubMed/NCBI

6 

Locatelli F, Nollke P, Zecca M, Korthof E, Lanino E, Peters C, Pession A, Kabisch H, Uderzo C, Bonfim CS, et al: Hematopoietic stem cell transplantation (HSCT) in children with juvenile myelomonocytic leukemia (JMML): Results of the EWOGMDS/EBMT trial. Blood. 105:410–419. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Locatelli F and Niemeyer CM: How I treat juvenile myelomonocytic leukemia. Blood. 125:1083–1090. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Wang PI and Marcotte EM: It's the machine that matters: Predicting gene function and phenotype from protein networks. J Proteomics. 73:2277–2289. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Hedges SB: The origin and evolution of model organisms. Nat Rev Genet. 3:838–849. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Helsmoortel HH: Gene expression profiling of 44 JMML patients and 7 healthy donors (discovery cohort). Homo sapiens. Dec 23–2015.

11 

Gautier L, Cope L, Bolstad BM and Irizarry RA: Affy-analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 20:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Bolstad BM, Irizarry RA, Astrand M and Speed TP: A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 19:185–193. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Smyth GK: Limma: Linear models for microarray data, in Bioinformatics and computational biology solutions using R and Bioconductor. Springer. 397–420. 2005.

15 

Liu H, Xu R, Liu X, Sun R and Wang Q: Bioinformatics analysis of gene expression in peripheralbloodmononuclearcells from children with type 1 diabetes in 3 periods. Exp Clin Endocrinol Diabetes. 122:477–483. 2014. View Article : Google Scholar : PubMed/NCBI

16 

da Huang W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2008. View Article : Google Scholar

17 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene Ontology: Tool for the unification of biology. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, Fridman WH, Pagès F, Trajanoski Z and Galon J: ClueGO: A Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 25:1091–1093. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, et al: STRING v10: Protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43:D447–D452. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Bandettini WP, Kellman P, Mancini C, Booker OJ, Vasu S, Leung SW, Wilson JR, Shanbhag SM, Chen MY and Arai AE: MultiContrast Delayed Enhancement (MCODE) improves detection of subendocardial myocardial infarction by late gadolinium enhancement cardiovascular magnetic resonance: A clinical validation study. J Cardiovasc Magn Reson. 14:832012. View Article : Google Scholar : PubMed/NCBI

23 

Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, et al: The Connectivity Map: Using gene-expression signatures to connect small molecules, genes, and disease. Science. 313:1929–1935. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Lamb J: The connectivity map: A new tool for biomedical research. Nat Rev Cancer. 7:54–60. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Zhang B, Kirov S and Snoddy J: WebGestalt: An integrated system for exploring gene sets in various biological contexts. Nucleic Acids Res. 33:W741–W748. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Chang TY, Dvorak CC and Loh ML: Bedside to bench in juvenile myelomonocytic leukemia: Insights into leukemogenesis from a rare pediatric leukemia. Blood. 124:2487–2497. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Sakashita K, Kato I, Daifu T, Saida S, Hiramatsu H, Nishinaka Y, Ebihara Y, Ma F, Matsuda K, Saito S, et al: In vitro expansion of CD34+CD38 cells under stimulation with hematopoietic growth factors on AGM-S3 cells in juvenile myelomonocytic leukemia. Leukemia. 29:606–614. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Gualtieri RJ, Castleberry RP, Gibbons J, Miller DM, Berkow RL, Parmley RT and Banks J: Cell culture studies and oncogene expression in juvenile chronic myelogenous leukemia. Exp Hematol. 16:613–619. 1988.PubMed/NCBI

29 

Gandre-Babbe S, Paluru P, Aribeana C, Chou ST, Bresolin S, Lu L, Sullivan SK, Tasian SK, Weng J, Favre H, et al: Patient-derived induced pluripotent stem cells recapitulate hematopoietic abnormalities of juvenile myelomonocytic leukemia. Blood. 121:4925–4929. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Hadland BK, Huppert SS, Kanungo J, Xue Y, Jiang R, Gridley T, Conlon RA, Cheng AM, Kopan R and Longmore GD: A requirement for Notch1 distinguishes 2 phases of definitive hematopoiesis during development. Blood. 104:3097–3105. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Kobayashi M, Shelley WC, Seo W, Vemula S, Lin Y, Liu Y, Kapur R, Taniuchi I and Yoshimoto M: Functional B-1 progenitor cells are present in the hematopoietic stem cell-deficient embryo and depend on Cbfβ for their development. Proc Natl Acad Sci USA. 111:12151–12156. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Lin Y, Yoder MC and Yoshimoto M: Lymphoid progenitor emergence in the murine embryo and yolk sac precedes stem cell detection. Stem Cells Dev. 23:1168–1177. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Yoshimoto M, Montecino-Rodriguez E, Ferkowicz MJ, Porayette P, Shelley WC, Conway SJ, Dorshkind K and Yoder MC: Embryonic day 9 yolk sac and intra-embryonic hemogenic endothelium independently generate a B-1 and marginal zone progenitor lacking B-2 potential. Proc Natl Acad Sci USA. 108:1468–1473. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Yoshimoto M, Porayette P, Glosson NL, Conway SJ, Carlesso N, Cardoso AA, Kaplan MH and Yoder MC: Autonomous murine T-cell progenitor production in the extra-embryonic yolk sac before HSC emergence. Blood. 119:5706–5714. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Mastrangelo MJ, Maguire HC Jr, Eisenlohr LC, Laughlin CE, Monken CE, McCue PA, Kovatich AJ and Lattime EC: Intratumoral recombinant gm-csf-encoding virus as gene therapy in patients with cutaneous melanoma. Cancer Gene Ther. 6:409–422. 1999. View Article : Google Scholar : PubMed/NCBI

36 

McNeel DG, Chen YH, Gulley JL, Dwyer AJ, Madan RA, Carducci MA and DiPaola RS: Randomized phase II trial of docetaxel with or without PSA-TRICOM vaccine in patients with castrate-resistant metastatic prostate cancer: A trial of the ECOG-ACRIN cancer research group (E1809). Hum Vaccin Immunother. 11:2469–2474. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Oudard S, Rixe O, Beuselinck B, Linassier C, Banu E, Machiels JP, Baudard M, Ringeisen F, Velu T, Lefrere-Belda MA, et al: A phase II study of the cancer vaccine TG4010 alone and in combination with cytokines in patients with metastatic renal clear-cell carcinoma: Clinical and immunological findings. Cancer Immunol Immunother. 60:261–271. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Ottolino-Perry K, Acuna SA, Angarita FA, Sellers C, Zerhouni S, Tang N and McCart JA: Oncolytic vaccinia virus synergizes with irinotecan in colorectal cancer. Mol. Oncol. 9:1539–1552. 2015.

39 

Quoix E, Lena H, Losonczy G, Forget F, Chouaid C, Papai Z, Gervais R, Ottensmeier C, Szczesna A, Kazarnowicz A, et al: TG4010 immunotherapy and first-line chemotherapy for advanced non-small-cell lung cancer (time): Results from the phase 2b part of a randomised, double-blind, placebo-controlled, phase 2B/3 trial. Lancet Oncol. 17:212–223. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Foy SP, Sennino B, dela Cruz T, Cote JJ, Gordon EJ, Kemp F, Xavier V, Franzusoff A, Rountree RB and Mandl SJ: Poxvirus-based active immunotherapy with PD-1 and LAG-3 dual immune checkpoint inhibition overcomes compensatory immune regulation, yielding complete tumor regression in mice. PLoS One. 11:e01500842016. View Article : Google Scholar : PubMed/NCBI

41 

Foy SP, Mandl SJ, dela Cruz T, Cote JJ, Gordon EJ, Trent E, Delcayre A, Breitmeyer J, Franzusoff A and Rountree RB: Poxvirus-based active immunotherapy synergizes with CTLA-4 blockade to increase survival in a murine tumor model by improving the magnitude and quality of cytotoxic T cells. Cancer Immunol Immunother. 65:537–549. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Cappuccini F, Stribbling S, Pollock E, Hill AV and Redchenko I: Immunogenicity and efficacy of the novel cancer vaccine based on simian adenovirus and MVA vectors alone and in combination with PD-1 mAb in a mouse model of prostate cancer. Cancer Immunol Immunother. 65:701–713. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Dimberg A: Chemokines in angiogenesis. Curr Top Microbiol Immunol. 341:59–80. 2010.PubMed/NCBI

44 

Speyer CL and Ward PA: Role of endothelial chemokines and their receptors during inflammation. J Invest Surg. 24:18–27. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Ben-Baruch A: The multifaceted roles of chemokines in malignancy. Cancer Metastasis Rev. 25:357–371. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Luther SA and Cyster JG: Chemokines as regulators of T cell differentiation. Nat Immunol. 2:102–107. 2001. View Article : Google Scholar : PubMed/NCBI

47 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr and Kinzler KW: Cancer genome landscapes. Science. 339:1546–1558. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Cancer Genome Atlas Research Network, . Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C and Stuart JM: The cancer genome atlas pan-cancer analysis project. Nat Genet. 45:1113–1120. 2013. View Article : Google Scholar : PubMed/NCBI

50 

International Cancer Genome Consortium, . Hudson TJ, Anderson W, Artez A, Barker AD, Bell C, Bernabé RR, Bhan MK, Calvo F, Eerola I, Gerhard DS, et al: International network of cancer genome projects. Nature. 464:993–998. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Braun S, Vogl FD, Naume B, Janni W, Osborne MP, Coombes RC, Schlimok G, Diel IJ, Gerber B, Gebauer G, et al: A pooled analysis of bone marrow micrometastasis in breast cancer. N Engl J Med. 353:793–802. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Janni W, Vogl FD, Wiedswang G, Synnestvedt M, Fehm T, Juckstock J, Borgen E, Rack B, Braun S, Sommer H, et al: Persistence of disseminated tumor cells in the bone marrow of breast cancer patients predicts increased risk for relapse-a European pooled analysis. Clin Cancer Res. 17:2967–2976. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Budowle B, Allard MW, Wilson MR and Chakraborty R: Forensics and mitochondrial DNA: Applications, debates, and foundations. Ann Rev Genom Hum Genet. 4:119–141. 2003. View Article : Google Scholar

54 

International HapMap Consortium, . Frazer KA, Ballinger DG, Cox DR, Hinds DA, Stuve LL, Gibbs RA, Belmont JW, Boudreau A, Hardenbol P, Leal SM, et al: A second generation human haplotype map of over 3.1 million SNPs. Nature. 449:851–861. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Cheng X, Chen G and Rodriguez WR: Micro-and nanotechnology for viral detection. Anal Bioanal Chem. 393:487–501. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Pinkel D, Segraves R, Sudar D, Clark S, Poole I, Kowbel D, Collins C, Kuo WL, Chen C, Zhai Y, et al: High resolution analysis of DNA copy number variation using comparative genomic hybridization to microarrays. Nat Genet. 20:207–211. 1998. View Article : Google Scholar : PubMed/NCBI

57 

Sozzi G, Conte D, Mariani L, Lo Vullo S, Roz L, Lombardo C, Pierotti MA and Tavecchio L: Analysis of circulating tumor DNA in plasma at diagnosis and during follow-up of lung cancer patients. Cancer Res. 61:4675–4678. 2001.PubMed/NCBI

58 

Bhowmick NA, Neilson EG and Moses HL: 2004. Stromal fibroblasts in cancer initiation and progression. Nature. 432:332–337. 2004. View Article : Google Scholar : PubMed/NCBI

59 

Sternlicht MD, Lochter A, Sympson CJ, Huey B, Rougier JP, Gray JW, Pinkel D, Bissell MJ and Werb Z: The stromal proteinase MMP3/stromelysin-1 promotes mammary carcinogenesis. Cell. 98:137–146. 1999. View Article : Google Scholar : PubMed/NCBI

60 

Paszek MJ, Zahir N, Johnson KR, Lakins JN, Rozenberg GI, Gefen A, Reinhart-King CA, Margulies SS, Dembo M, Boettiger D, et al: Tensional homeostasis and the malignant phenotype. Cancer Cell. 8:241–254. 2005. View Article : Google Scholar : PubMed/NCBI

61 

Erler JT, Bennewith KL, Nicolau M, Dornhöfer N, Kong C, Le QT, Chi JT, Jeffrey SS and Giaccia AJ: Lysyl oxidase is essential for hypoxia-induced metastasis. Nature. 440:1222–1226. 2006. View Article : Google Scholar : PubMed/NCBI

62 

Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SF, Csiszar K, Giaccia A, Weninger W, et al: Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 139. 1–906. 2009.

63 

Hynes RO: The extracellular matrix: Not just pretty fibrils. Science. 326:1216–1219. 2009. View Article : Google Scholar : PubMed/NCBI

64 

Wiseman BS, Sternlicht MD, Lund LR, Alexander CM, Mott J, Bissell MJ, Soloway P, Itohara S and Werb Z: Site-specific inductive and inhibitory activities of MMP-2 and MMP-3 orchestrate mammary gland branching morphogenesis. J Cell Biol. 162:1123–1133. 2003. View Article : Google Scholar : PubMed/NCBI

65 

Stickens D, Behonick DJ, Ortega N, Heyer B, Hartenstein B, Yu Y, Fosang AJ, Schorpp-Kistner M, Angel P and Werb Z: Altered endochondral bone development in matrix metalloproteinase 13-deficient mice. Development. 131:5883–5895. 2004. View Article : Google Scholar : PubMed/NCBI

66 

Rebustini IT, Myers C, Lassiter KS, Surmak A, Szabova L, Holmbeck K, Pedchenko V, Hudson BG and Hoffman MP: MT2-MMP-dependent release of collagen IV NC1 domains regulates submandibular gland branching morphogenesis. Dev Cell. 17:482–493. 2009. View Article : Google Scholar : PubMed/NCBI

67 

Lu P, Takai K, Weaver VM and Werb Z: Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb Perspect Biol. 3:a0050582011. View Article : Google Scholar : PubMed/NCBI

68 

Cox TR and Erler JT: Remodeling and homeostasis of the extracellular matrix: Implications for fibrotic diseases and cancer. Dis Model Mech. 4:165–178. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Kitamura T, Qian BZ and Pollard JW: Immune cell promotion of metastasis. Nat Rev Immunol. 15:73–86. 2015. View Article : Google Scholar : PubMed/NCBI

70 

Grivennikov SI, Greten FR and Karin M: Immunity, inflammation, and cancer. Cell. 140:883–899. 2010. View Article : Google Scholar : PubMed/NCBI

71 

DeNardo DG and Coussens LM: Inflammation and breast cancer. Balancing immune response: Crosstalk between adaptive and innate immune cells during breast cancer progression. Breast Cancer Res. 9:2122007. View Article : Google Scholar : PubMed/NCBI

72 

Hatzoglou A, Ouafik L, Bakogeorgou E, Thermos K and Castanas E: Morphine cross-reacts with somatostatin receptor SSTR2 in the T47D human breast cancer cell line and decreases cell growth. Cancer Res. 55:5632–5636. 1995.PubMed/NCBI

73 

Rasmussen M, Zhu W, Tønnesen J, Cadet P, Tønnesen E and Stefano GB: Effects of morphine on tumor growth. Neuro Endocrinol Lett. 23:193–198. 2002.PubMed/NCBI

74 

Mayer TU, Kapoor TM, Haggarty SJ, King RW, Schreiber SL and Mitchison TJ: Small molecule inhibitor of mitotic spindle bipolarity identified in a phenotype-based screen. Science. 286:971–974. 1999. View Article : Google Scholar : PubMed/NCBI

75 

Crews CM and Mohan R: Small-molecule inhibitors of the cell cycle. Curr Opin Chem Biol. 4:47–53. 2000. View Article : Google Scholar : PubMed/NCBI

76 

Liu X, Gong H and Huang K: Oncogenic role of kinesin proteins and targeting kinesin therapy. Cancer Sci. 104:651–656. 2013. View Article : Google Scholar : PubMed/NCBI

77 

Kapoor TM, Mayer TU, Coughlin ML and Mitchison TJ: Probing spindle assembly mechanisms with monastrol, a small molecule inhibitor of the mitotic kinesin, Eg5. J Cell Biol. 150:975–988. 2000. View Article : Google Scholar : PubMed/NCBI

78 

DeBonis S, Simorre JP, Crevel I, Lebeau L, Skoufias DA, Blangy A, Ebel C, Gans P, Cross R, Hackney DD, et al: Interaction of the mitotic inhibitor monastrol with human kinesin Eg5. Biochemistry. 42:338–349. 2003. View Article : Google Scholar : PubMed/NCBI

79 

Cochran JC and Gilbert SP: ATPase mechanism of Eg5 in the absence of microtubules: Insight into microtubule activation and allosteric inhibition by monastrol. Biochemistry. 44:16633–16648. 2005. View Article : Google Scholar : PubMed/NCBI

80 

Haque SA, Hasaka TP, Brooks AD, Lobanov PV and Baas PW: Monastrol, a prototype anti-cancer drug that inhibits a mitotic kinesin, induces rapid bursts of axonal outgrowth from cultured postmitotic neurons. Cell Motil Cytoskeleton. 58:10–16. 2004. View Article : Google Scholar : PubMed/NCBI

81 

Maliga Z, Kapoor TM and Mitchison TJ: Evidence that monastrol is an allosteric inhibitor of the mitotic kinesin Eg5. Chem Biol. 9:989–996. 2002. View Article : Google Scholar : PubMed/NCBI

82 

Müller C, Gross D, Sarli V, Gartner M, Giannis A, Bernhardt G and Buschauer A: Inhibitors of kinesin Eg5: Antiproliferative activity of monastrol analogues against human glioblastoma cells. Cancer Chemother Pharmacol. 59:157–164. 2007. View Article : Google Scholar : PubMed/NCBI

83 

Cascinu S, Bichisao E, Amadori D, Silingardi V, Giordani P, Sansoni E, Luppi G, Catalano V, Agostinelli R and Catalano G: High-dose loperamide in the treatment of 5-fluorouracil-induced diarrhea in colorectal cancer patients. Support Care Cancer. 8:65–67. 2000.PubMed/NCBI

84 

Vail DM: Supporting the veterinary cancer patient on chemotherapy: Neutropenia and gastrointestinal toxicity. Top Companion Anim Med. 24:122–129. 2009. View Article : Google Scholar : PubMed/NCBI

85 

Gong XW, Xu YH, Chen XL and Wang YX: Loperamide, an antidiarrhea drug, has antitumor activity by inducing cell apoptosis. Pharmacol Res. 65:372–378. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Ohman R and Axelsson R: Relationship between prolactin response and antipsychotic effect of thioridazine in psychiatric-patients. Eur J Clin Pharmacol. 14:111–116. 1978. View Article : Google Scholar : PubMed/NCBI

87 

Realmuto GM, Erickson WD, Yellin AM, Hopwood JH and Greenberg LM: Clinical comparison of thiothixene and thioridazine in schizophrenic adolescents. Am J Psychiatry. 141:440–442. 1984. View Article : Google Scholar : PubMed/NCBI

88 

van Soolingen D, Hernandez-Pando R, Orozco H, Aguilar D, Magis-Escurra C, Amaral L, van Ingen J and Boeree MJ: The antipsychotic thioridazine shows promising therapeutic activity in a mouse model of multidrug-resistant tuberculosis. PLoS One. 5:e126402010. View Article : Google Scholar : PubMed/NCBI

89 

Thorsing M, Klitgaard JK, Atilano ML, Skov MN, Kolmos HJ, Filipe SR and Kallipolitis BH: Thioridazine induces major changes in global gene expression and cell wall composition in methicillin-resistant Staphylococcus aureus USA300. PLoS One. 8:e645182013. View Article : Google Scholar : PubMed/NCBI

90 

Byun HJ, Lee JH, Kim BR, Kang S, Dong SM, Park MS, Lee SH, Park SH and Rho SB: Anti-angiogenic effects of thioridazine involving the FAK-mTOR pathway. Microvasc Res. 84:227–234. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Park MS, Dong SM, Kim BR, Seo SH, Kang S, Lee EJ, Lee SH and Rho SB: Thioridazine inhibits angiogenesis and tumor growth by targeting the VEGFR-2/PI3K/mTOR pathway in ovarian cancer xenografts. Oncotarget. 5:4929–4934. 2014. View Article : Google Scholar : PubMed/NCBI

92 

Kang S, Dong SM, Kim BR, Park MS, Trink B, Byun HJ and Rho SB: Thioridazine induces apoptosis by targeting the PI3K/Akt/mTOR pathway in cervical and endometrial cancer cells. Apoptosis. 17:989–997. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Csonka Á, Spengler G, Martins A, Ocsovszki I, Christensen JB, Hendricks O, Kristiansen JE, Amaral L and Molnar J: Effect of thioridazine stereoisomers on the drug accumulation of mouse lymphoma and human prostate cancer cell lines in vitro. In Vivo. 27:815–820. 2013.PubMed/NCBI

94 

Mu J, Xu H, Yang Y, Huang W, Xiao J, Li M, Tan Z, Ding Q, Zhang L, Lu J, et al: Thioridazine, an antipsychotic drug, elicits potent antitumor effects in gastric cancer. Oncol Rep. 31:2107–2114. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Nagel D, Spranger S, Vincendeau M, Grau M, Raffegerst S, Kloo B, Hlahla D, Neuenschwander M, von Kries Peter J, Hadian K, et al: Pharmacologic inhibition of MALT1 protease by phenothiazines as a therapeutic approach for the treatment of aggressive ABC-DLBCL. Cancer Cell. 22:825–837. 2012. View Article : Google Scholar : PubMed/NCBI

96 

Sachlos E, Risueño RM, Laronde S, Shapovalova Z, Lee JH, Russell J, Malig M, McNicol JD, Fiebig-Comyn A, Graham M, et al: Identification of drugs including a dopamine receptor antagonist that selectively target cancer stem cells. Cell. 149:1284–1297. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Ke XY, Lin Ng VW, Gao SJ, Tong YW, Hedrick JL and Yang YY: Co-delivery of thioridazine and doxorubicin using polymeric micelles for targeting both cancer cells and cancer stem cells. Biomaterials. 35:1096–1108. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Nudelman A, Ruse M, Aviram A, Rabizadeh E, Shaklai M, Zimrah Y and Rephaeli A: Novel anticancer prodrugs of butyric acid. 2. J Med Chem. 35:687–694. 1992. View Article : Google Scholar : PubMed/NCBI

99 

Wu CH, Jeng JH, Wang YJ, Tseng CJ, Liang YC, Chen CH, Lee HM, Lin JK, Lin CH, Lin SY, et al: Antitumor effects of miconazole on human colon carcinoma xenografts in nude mice through induction of apoptosis and G0/G1 cell cycle arrest. Toxicol Appl Pharmacol. 180:22–35. 2002. View Article : Google Scholar : PubMed/NCBI

100 

Lewinska A, Adamczyk-Grochala J, Kwasniewicz E, Deregowska A and Wnuk M: Ursolic acid-mediated changes in glycolytic pathway promote cytotoxic autophagy and apoptosis in phenotypically different breast cancer cells. Apoptosis. 22:800–815. 2017. View Article : Google Scholar : PubMed/NCBI

101 

Jiao Y, Hannafon BN, Zhang RR, Fung KM and Ding WQ: Docosahexaenoic acid and disulfiram act in concert to kill cancer cells: A mutual enhancement of their anticancer actions. Oncotarget. 8:17908–17920. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 40 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao W, Wang L and Yu Y: Gene module analysis of juvenile myelomonocytic leukemia and screening of anticancer drugs. Oncol Rep 40: 3155-3170, 2018
APA
Zhao, W., Wang, L., & Yu, Y. (2018). Gene module analysis of juvenile myelomonocytic leukemia and screening of anticancer drugs. Oncology Reports, 40, 3155-3170. https://doi.org/10.3892/or.2018.6709
MLA
Zhao, W., Wang, L., Yu, Y."Gene module analysis of juvenile myelomonocytic leukemia and screening of anticancer drugs". Oncology Reports 40.6 (2018): 3155-3170.
Chicago
Zhao, W., Wang, L., Yu, Y."Gene module analysis of juvenile myelomonocytic leukemia and screening of anticancer drugs". Oncology Reports 40, no. 6 (2018): 3155-3170. https://doi.org/10.3892/or.2018.6709