You are on page 1of 10

Computers in Biology and Medicine 134 (2021) 104470

Contents lists available at ScienceDirect

Computers in Biology and Medicine


journal homepage: www.elsevier.com/locate/compbiomed

New drug candidates for osteosarcoma: Drug repurposing based on gene


expression signature
Raissa Coelho Andrade a, b, Mariana Boroni c, d, Marion Kielmanowicz Amazonas a,
Fernando Regla Vargas a, b, *
a
Birth Defects Epidemiology Laboratory, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
b
Genetics and Molecular Biology Department, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
c
Bioinformatics and Computational Biology Lab, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
d
Experimental Medicine Research Cluster (EMRC), University of Campinas (UNICAMP), Campinas, Brazil

A R T I C L E I N F O A B S T R A C T

Keywords: Osteosarcoma (OS) is an aggressive bone malignancy and the third most common cancer in adolescence. Since
Osteosarcoma the late 1970s, OS therapy and prognosis had only modest improvements, making it appealing to explore new
Microarray expression data tools that could help ameliorate the treatment. We present a meta-analysis of the gene expression signature of
Gene expression signature
primary OS, and propose small molecules that could reverse this signature. The meta-analysis was performed
Drug repurposing
Afatinib
using GEO microarray series. We first compared gene expression from eleven primary OS against osteoblasts to
obtain the differentially expressed genes (DEGs). We later filtered those DEGs by verifying which ones had a
concordant direction of differential expression in a validation group of 82 OS samples versus 30 bone marrow
mesenchymal stem cells (BM-MSC) samples. A final gene expression signature of 266 genes (98 up and 168 down
regulated) was obtained. The L1000CDS2 engine was used for drug repurposing. The top molecules predicted to
reverse the signature were afatinib (PubChem CID 10184653), BRD-K95196255 (PubChem CID 3242434), DG-
041 (PubChem CID 11296282) and CA-074 Me (PubChem CID 23760717). Afatinib (Gilotrif™) is currently used
for metastatic non-small-cell lung cancer with EGFR mutations, and in vitro evidence shows antineoplastic po­
tential in OS cells. The other three molecules have reports of antineoplastic effects, but are not currently FDA-
approved. Further studies are necessary to establish the potential of these drugs in OS treatment. We believe
our results can be an important contribution for the investigation of new therapeutic genetic targets and for
selecting new drugs to be tested for OS.

1. Introduction malignancy [4].


In adolescents and young adults, the most common sites of OS are the
Osteosarcoma (OS) is an aggressive bone malignancy that typically metaphysis of the distal femur, the proximal tibia and the proximal
occurs in the second decade of life, during the pubertal growth spurt [1]. humerus, with the most common clinical presentation being chronic
It is the third most common cancer in adolescence, with approximately pain with indolent evolution in the affected area [1]. Prognosis of OS has
60% of the cases occurring before the age of 25 years old [2,3]. A sec­ significantly improved with the introduction of chemotherapy in the late
ondary incidence peak is observed in patients over 65 years old, 1970s, but since then, therapy has remained essentially the same [5–7],
although in these cases it is most commonly associated with a previous and the current 5-year overall survival is still less than 70% in localized
bone disease, while in young individuals it is considered a primary disease, and less than 20% in 3 years for metastatic disease, that occurs

Abbreviations: BM-MSC, bone marrow mesenchymal stromal/stem cells; DEGs, Differentially expressed genes; ECM, extracellular matrix; FDR, false discovery
rate; GEO, Gene Expression Omnibus (database); GSE, GEO series; GSEA, Gene Set Enrichment Analysis; KEGG, Kyoto Encyclopedia of Genes and Genomes;
L1000CDS2, LINCS L1000 characteristic direction signatures; ORA, Overrepresentation analysis; OS, Osteosarcoma; WebGestalt, WEBbased GEne SeT AnaLysis
Toolkit.
* Corresponding author. Birth Defects Epidemiology Laboratory, Oswaldo Cruz Foundation (FIOCRUZ), Av. Brasil 4365 – Pavilhão Leônidas Deane Sala 617,
Manguinhos, Rio de Janeiro, 21040-360, Brazil.
E-mail address: fernando.vargas@ioc.fiocruz.br (F.R. Vargas).

https://doi.org/10.1016/j.compbiomed.2021.104470
Received 25 February 2021; Received in revised form 1 May 2021; Accepted 2 May 2021
Available online 7 May 2021
0010-4825/© 2021 Elsevier Ltd. All rights reserved.
R.C. Andrade et al. Computers in Biology and Medicine 134 (2021) 104470

most commonly in the lungs [7]. Surgery is the pillar of the treatment, samples extracted and processed in loco, excluding studies that used
associated to neoadjuvant and adjuvant chemotherapy. Four chemo­ purchased BM-MSCs. We also excluded studies that used BM-MSCs
therapy agents are included in the standard protocols as first line extracted from patients with osteoarthritis or osteoporosis.
treatment: methotrexate with leucovorin rescue, doxorubicin, cisplatin,
and ifosfamide [2]. 2.2. Data processing and differential gene expression
The etiology of OS remains obscure. Risk factors for the young pa­
tients include taller stature, rapid bone growth, male sex, high birth The analysis were performed with the statistical computing envi­
weight and some genetic conditions such as Li-Fraumeni syndrome, ronment R. Raw data from each of the selected GEO series (microarray.
hereditary retinoblastoma and syndromes related to mutations in the CEL files) were converted to expression measures and normalized using
RECQL family of genes (like Rothmund–Thomson syndrome) [1,8]. either affy or oligo packages [13,14]. For integrating datasets from
Previous exposure to ionizing radiation is the only recurrently reported different platforms, the probe IDs were converted to the respective Gene
environmental risk factor for the young [1,8]. Symbols, retrieved from the annotation package of each Affymetrix
Histologically, osteosarcomas are marked for the production of platform. When more than one probe for the same gene was present, the
osteoid tissue and the most common type is the conventional OS, rep­ mean expression value of the probes was calculated.
resenting >80% of the cases, and characterized by different amounts of Test group: the expression matrices generated by the two datasets in
osteoblastic, chondroblastic, and fibroblastic cells [2]. Two types of cells the test group were combined. Only the genes in common between the
have been suggested as the tumor cell-of-origin: mesenchymal stem cells two different Affymetrix platforms were kept for further analysis. The
and a more differentiated/committed osteoblastic cell population, both ComBat function (sva package [15]) was applied to the samples. This
of which could go through malignant transformation within the appro­ function removes the batch effect, allowing data from different experi­
priate molecular signals [9,10]. The typical genomic profile of OS is ments to be comparable in the same analysis, which increases the
described as chaotic and complex, including a high somatic mutation robustness of the results obtained. Then, differential gene expression
rate and kataegis, complex chromosomal rearrangements, inactivating analysis between osteosarcoma and osteoblasts was conducted using the
mutations in TP53 and RB1, and activating mutations in PI3K/Akt/m­ Limma package [16]. For the identification of differentially expressed
TOR pathway genes [11]. Commonly gene expression alterations genes in microarray data, a combination of thresholds for adjusted
include the Notch and Wnt pathway upregulation, overexpression of p-value (FDR) and log fold change yields better results than just the
Runx2 and Osterix (transcription factors required for osteoblast differ­ p-value or fold change alone [17]. In this study, DEGs were defined by
entiation), EZR (responsible for linking the cytoskeleton to the cell applying a log fold change (logFC) threshold of >2, and a false discovery
membrane), downregulation of Fas and Fas ligand (regulators of cellular rate (FDR) threshold of <0.05, which are common thresholds for this
apoptosis) and receptor tyrosine kinase genes like ALX and HER-2 [1, analysis [18–22].
12]. Validation group: as in the test group, the expression matrices of the
Despite the many studies developed to decode the molecular causes eight datasets in the validation group were combined, genes that were
of OS, translational applicability has remained a challenge. The insuf­ not present in all the different Affymetrix platforms were filtered out,
ficient improvement in chemotherapy regimens and survival over the and the ComBat function was applied to the samples. Except that here,
last decades makes it appealing to explore new tools for discovering each dataset included exclusively tumor or normal samples. Therefore,
small molecules that could help ameliorate OS treatment [7]. In this since the ComBat function would also remove the expression differences
study, we present a meta-analysis of the gene expression signature from caused by the biological status of the samples, we applied the function in
primary OS tumors, and propose small molecules that could reverse this two steps: first in the tumor samples, then in the normal samples. To
signature, based on a drug repurposing bioinformatics tool. We believe minimize the possible biases inserted by this approach, we did not use
the results presented here can serve as a groundwork for further inves­ the same statistical criteria applied to the test group to define differen­
tigation targeting OS altered pathways. tially expressed genes. Instead, after performing the analysis with
Limma, we explored the expression consistency of the DEGs identified in
2. Methods the test group by verifying if these genes had a concordant direction of
differential expression in the validation group. The genes with a
2.1. Data collection concordant differential expression were considered validated and
referred to as the gene expression signature for OS. Heat maps were
We searched the GEO database (www.ncbi.nlm.nih.gov/geo/) to constructed considering the OS gene signature for both the test and
retrieve different series of samples that accessed the mRNA expression validation groups using the pheatmap package version 1.0.12. Genes
profiles in osteosarcoma and normal bone samples. Our filters included: (rows) in the heatmap of the test group were clustered using the 1-Pear­
organism: “Homo sapiens” and study type: “Expression profiling by son correlation coefficient as the distance metrics and “average” as the
array”, focused in the Affymetrix platforms. The search terms included agglomeration method; in the heatmap of the validation group, the
“osteosarcoma”, “bone marrow mesenchymal stromal/stem cells” (or genes were arranged in the same order of the test group. The samples
“BM-MSC”), and “osteoblasts”. For the OS samples, we only considered (columns) were also clustered using the 1-Pearson correlation coeffi­
studies that used primary specimens, i.e.: biopsies and surgical samples, cient as the distance metrics and “average” as the agglomeration method
excluding studies that used cell lines derived from tumor tissue. OS for both the heatmaps.
samples from metastases were also excluded. For all the data series,
median age of the patients and healthy donors should be < 40 years old, 2.3. Functional analysis
and the number of relevant samples in each series should be ≥ 3. Other
tissue types investigated in each series were not included in the analysis. The differential expression analysis of the test group was assessed for
Our analysis was divided into two steps: test group and validation group. enriched pathways using the Gene Set Enrichment Analysis (GSEA),
Test group: we searched for series that included both tumor and with the respective log fold change scores for each gene. Then, the
normal samples in the same experiment, to properly remove the batch validated gene signature was assessed for enriched pathways through
effect while performing the meta-analysis. the overrepresentation analysis (ORA) methodology. Both in­
Validation group: we validated the results using independent series vestigations were performed in the WEBbased GEne SeT AnaLysis
that included either primary tumor tissues from patients with OS or BM- Toolkit (WebGestalt) using the Kyoto Encyclopedia of Genes and Ge­
MSC samples from healthy controls. For the BM-MSC samples, we only nomes (KEGG) database [23]. The p-values were controlled for false
considered studies that used early passage MSCs from bone marrow discovery rate (FDR) using the Bonferroni method, considered

2
R.C. Andrade et al. Computers in Biology and Medicine 134 (2021) 104470

significant when <0.05. down-regulated in OS (Fig. 1A). We then assessed the expression con­
sistency of these DEGs by verifying if they had a concordant direction of
2.4. Drug repurposing differential expression in the validation group (i.e.: same signed logFC).
Of the 283 genes, 280 had matched gene symbols across all the different
The gene signature obtained was used as an input in the LINCS L1000 Affymetrix array platforms used in the validation groups (106 of the up-
characteristic direction signatures search engine (L1000CDS2) [24] to regulated and 174 of the down-regulated genes). These 280 genes
search for drugs and small molecules that could reverse the expression yielded a 95% (266/280) differential expression concordance rate be­
signature of OS samples. This engine is part of the Library of Integrated tween the test and validation groups: 98/106 up-regulated genes and
Network-based Cellular Signatures (LINCS) project, which investigates 168/174 down-regulated genes (Fig. 1B). The heatmaps comparing the
expression profiles generated by small molecules (perturbagens) in expression patterns of these 280 genes between test and validation
human cells. The gene expression perturbations in the LINCS mRNA groups are exhibited in Fig. 1C. The top 20 up and down-regulated genes
profiling assay come from tests with multiple cell lines, mostly observed in the test group are shown in box 1. The full list with the up
cancerous ones, including the U2OS cell line, originated from osteo­ and down-regulated genes is available at Supplementary Tables 1 and 2
sarcoma [24,25]. When up/down regulated gene lists are submitted to
L1000CDS2, they are compared to the genes computed from the LINCS 3.3. Functional analysis
L1000 data and the top 50 molecules which either mimic or reverse
(according to the user’s selection) the input expression profile are Gene set enrichment analysis for KEGG pathways in the test group is
returned, with a score for each drug. The score is the number of over­ represented in Fig. 2A and Table 2. The over-representation analysis,
lapping genes between the input and the database signature for the drug performed with the subset of 98 up- and the 168 down-regulated vali­
divided by the effective input (i.e., the genes from the input that are dated genes is exhibited in Fig. 2B (top and bottom graphics, respec­
present in the L1000 database). The top results were manually refer­ tively). Positively enriched pathways were associated to inflammatory
enced in the database PubChem [26] for further analysis of the drugs of response, while negatively enriched pathways included p53-signaling.
interest. The ORA analysis revealed additional down-regulated pathways such
as extracellular matrix (ECM) receptor interaction, proteoglycans in
3. Results cancer and focal adhesion. The concomitant pathways between the
GSEA and ORA analysis are shown in bold in Table 2.
3.1. Selected data
3.4. Drug repositioning in OS
Our selection criteria led us to 10 independent GEO series, charac­
terized in Table 1. The test group was composed by GSE12865 and The LINCS L1000 Characteristic Direction Signature Search Engine
GSE14359, which included, together, eleven primary OS tumor samples was utilized to query the best molecules which could reverse the gene
in duplicate and two osteoblast cell lines in duplicate. The validation expression signature for OS (i.e., the validated differentially expressed
group included eight independent series: three series with a total of 82 genes), and thus be investigated for the treatment of this neoplasm.
primary tumor tissues from patients with OS (GSE14827, GSE16091, Fig. 2C exhibits the clustergram with the best 50 matches of small
GSE87437) and five series with a total of 30 BM-MSC samples from molecules, of which 40 were unique (the same molecule can have
healthy controls (GSE84881, GSE18698, GSE110359, GSE113736, multiple entries in the L1000CDS2 database, one for each cell line it was
GSE122778). tested). Four drug candidates were predicted to reverse the expression of

Table 1
Characteristics of the microarray GSE series used in this study.
GEO series Type of sample No of samples Naive tumor samples?a Median age of patients/controls (years) Affymetrix platform

Test group
GSE12865 Primary tumor/osteoblasts 6/1b Yes NA (all pediatric) HuGene-1_0-st
GSE14359 Primary tumor/osteoblasts 5/1b NA 17 HG-U133A
Validation group
GSE14827 Primary tumor 27 Yes 15 HG-U133_Plus_2
GSE16091 Primary tumor 34 Yes 12,5 HG-U133A
GSE87437 Primary tumor 21 Yes 15 HG-U133_Plus_2
GSE84881 BM-MSC 4 – 22,5 HG-U133_Plus_2
GSE18698 BM-MSC 3 – 36 HuEx-1_0-st
GSE110359 BM-MSC 5 – 14,6c HTA-2_0
GSE113736 BM-MSC 4d – 26,5 HuEx-1_0-st
GSE122778 BM-MSC 14 – 36 HuGene-1_0-st

NA: non-available information. BM-MSC: Bone marrow mesenchymal stem cells.


a
Naive tumor samples refer to tumors collected before chemotherapy. b Number of original samples. The duplicates available in GEO were also used. c Mean age
(median age not available)d Only the first replicate of each sample of GSE113736 was used.

3.2. Differential gene expression between osteosarcoma and normal bone at least 10% of our input genes. Of these molecules, only one is currently
samples FDA-approved: afatinib (S1011; PubChem CID 10184653), which was
also the best scored match in our search, and is already used as an
We searched for differentially expressed genes in osteosarcoma by antineoplastic agent. The gene modulation of afatinib in OS predicted by
comparing gene expression levels between osteosarcoma samples and our analysis is exhibited in Table 3. Besides afatinib, three molecules
osteoblasts in our test group. A total of 12,040 genes were analyzed. met our selection criteria (Table 3): BRD-K95196255 (PubChem CID
After applying a log fold change threshold of >2 and an FDR threshold of 3242434), which is a hydroxyquinoline with cytotoxic activity; DG-041
<0.05, 283 genes were identified: 108 were up-regulated and 175 were (or DTSI; PubChem CID 11296282), that acts as a prostaglandin receptor

3
R.C. Andrade et al. Computers in Biology and Medicine 134 (2021) 104470

Fig. 1. Identification of OS gene signature. A) Volcano plot of the gene expression profile identified in the test group. Thresholds for DEGs were log fold change
>2/< − 2 and FDR <0.05. B) Diagram evidencing the consistency of expression direction of these genes between the test and validation groups. C) Heatmaps of DEGs
in test and validation groups. Columns correspond to the samples and rows correspond to the genes. Each batch represents one GEO series. Left: Heatmap of the test
group, exhibiting the expression patterns of 280 DEGs between 11 primary OS samples and two osteoblast cell lines. Batch 1: GSE14359; batch 2: GSE12865. Right:
Heatmap of the validation group, exhibiting the expression patterns of 280 DEGs between 82 primary OS samples and 30 BM-MSC samples. Batch 1: GSE14827; batch
2: GSE16091; batch 3: GSE84881; batch 4: GSE87437; batch 5: GSE110359; batch 6: GSE113736; batch 7: GSE122778; batch 8: GSE18698. The concordance rate of
direction of expression (i.e.: same-signed logFC) between test and validation group was 95% (266 of 280 genes).

4
R.C. Andrade et al. Computers in Biology and Medicine 134 (2021) 104470

Box 1
Top up and down-regulated genes in OS. LogFC and FDR are correspondent to the test group.

Top 20 up and down-regulated genes in OS


Up-regulated genes Down-regulated genes
Gene symbol logFC FDR Gene symbol logFC FDR
MDFI 2.556284 2.767355e-12 TPD52L1 − 3.338841 6.711174e-17
A2M 5.909319 2.304380e-11 PENK − 4.364210 8.209018e-16
RNF144A 2.140703 5.632540e-10 SYNC − 3.262922 1.739980e-15
ARHGDIB 3.775558 1.039917e-09 FGF2 − 3.480656 2.936067e-15
MCAM 2.426295 4.577510e-09 GREM2 − 2.648890 1.134880e-14
ALPL 4.137270 8.122120e-09 LRRC2 − 2.798352 1.260484e-14
LCP1 2.659893 9.478168e-09 SLC14A1 − 4.672709 1.260484e-14
CPVL 3.240216 3.238323e-08 FGF5 − 4.050403 4.140875e-14
LYZ 4.689291 3.847037e-08 ISLR − 3.886011 5.889290e-14
HLA-DRA 5.400430 5.187442e-08 PHLDA3 − 2.310864 1.324531e-13
RASGRP3 2.216738 1.275734e-07 IL26 − 2.670094 2.534270e-13
CD93 2.396420 1.678738e-07 AVPI1 − 2.987813 2.895176e-13
HLA-DRB1 3.936381 2.185956e-07 RPS6KA2 − 3.129784 4.639098e-13
PECAM1 3.413068 2.585345e-07 QSOX1 − 3.214736 8.528371e-13
LAPTM5 3.777067 3.172885e-07 MOXD1 − 2.830525 1.025251e-12
CD53 3.143652 4.841996e-07 SERPINE1 − 4.864428 1.226166e-12
ELMO1 2.538471 8.119163e-07 AOX1 − 2.199167 1.285913e-12
GPC4 2.459877 1.500226e-06 CITED2 − 3.383479 1.584868e-12
CPE 4.395745 1.535049e-06 VGLL3 − 3.279918 1.584868e-12
MEF2C 3.308009 1.584478e-06 BST1 − 2.245540 2.041588e-12

antagonist; and CA-074 methyl ester (PubChem CID 23760717), which been a debate over which is the cell of origin of osteosarcoma. The two
is a cathepsin inhibitor. The full list of molecules is available in Sup­ best candidates are mesenchymal stem cells and a more differentiated/
plementary Table 3. committed osteoblastic cell population [9,10]. The primary location of
OS within metaphysis of long bones and its peak occurrence during rapid
4. Discussion growth phases in adolescence imply that rapidly dividing cells are
involved in this process. Both mesenchymal stem cells and
Osteosarcoma is an aggressive primary malignancy of the bone, with pre-osteoblasts fit this requirement, and there are in vitro and in vivo
very limited information regarding environmental risk factors associ­ evidences in favor of both cell types as possible cells of origin [9,10]. The
ated to its etiology [27,28]. Also, due to its rarity and peak incidence in most likely scenario is that either cell type may originate OS under the
adolescence, osteosarcoma may often be missed on an initial physical influence of proper genetic background, microenvironmental or epige­
exam, since the primary symptom of chronic pain may be wrongly netic signals [9]. Indeed, the results obtained here show a concordant
attributed to athletics or rapid bone growth [27,29]. Those factors may direction of differential expression of OS DEGs when compared both to
difficult the development of prevention and early detection strategies. osteoblasts and BM-MSCs, favoring this hypothesis.
Indeed, since the mid-1980s, only modest progress has been made Also importantly, our analysis only considered primary tumor sam­
concerning the survival of patients diagnosed with osteosarcoma [5]. ples for evaluation, excluding OS cell lines, which can eliminate false
Currently, the 5-year event-free survival of patients is 54%, even if positive results due to differences in expression pattern between tumor
complete surgical resection is possible at diagnosis [6]. A better un­ and tumor cell line samples [31]. Likewise, we selected series of patients
derstanding of OS molecular features may enhance the development of with a median age <40 years. It is known that OS has two peak in­
therapeutic strategies and have an impact on the morbidity and out­ cidences through life: adolescents/young adults and elderly [27]. In the
comes for this disease [30]. elderly, OS often arises in the context of a previous condition, such as the
In this study, we searched for a gene expression signature for OS, by malignant transformation of Paget disease of bone or post radiotherapy
performing a meta-analysis from different GEO microarray series. We [32]. Thus, our selection criteria can give a more trustworthy expression
therefore managed to analyze a large number of samples of a rare tumor profile of the primary OS tumors of young individuals. Another advan­
while also applying stringent selection criteria to the data. As a first step, tage is that at least 88 of the 93 OS samples analyzed in this study were
we compared gene expression from eleven primary OS samples against naive samples, avoiding possible alterations in gene expression that
osteoblasts to obtain the significantly differentially expressed genes. We could be caused by chemotherapeutic treatment.
later filtered those DEGs by verifying which ones had a concordant di­ Among the DEGs identified in our analysis MDFI was the most up-
rection of differential expression in a validation group of 82 primary OS regulated gene. This gene encodes the MDFI protein, a MyoD family
samples versus 30 BM-MSC samples from healthy controls. Our analysis inhibitor [33], thus inhibiting myogenesis differentiation of mesen­
yielded a 95% differential expression concordance rate between the test chymal lineage cells. MyoD can upregulate expression of p21 and
and validation groups, with a final number of 266 genes (98 up and 168 maintain the tumor suppressor pRb in its active state. Thus, MyoD both
down-regulated) deemed to constitute the gene expression signature. induces myogenic phenotype and promotes cell cycle arrest, which
An important approach in this work was the use of different cell types emphasizes the interconnection between cell differentiation and growth
as normal bone controls in the test and validation groups. There has arrest [34]. The disruption of these mechanisms by MDFI up-regulation

5
R.C. Andrade et al. Computers in Biology and Medicine 134 (2021) 104470

Fig. 2. Enriched pathways for OS and drugs predicted to reverse OS signature. A) GSEA analysis for the OS test group. Top 10 up-regulated KEGG pathways (in
blue) and top 10 down-regulated KEGG pathways (in orange). B) Over-representation (ORA) pathway analysis of the 266 validated genes. The top 10 up-regulated
pathways (top graphic) and the top 10 down-regulated pathways (bottom graphic). C) Clustergram with the top ranked L1000 molecules predicted to reverse the OS
gene expression signature. Columns include the best ranked drugs, in descending order. Each blue cell represents an up-regulated gene in OS which is downregulated
by the respective drug in the column. Each red cell represents a down-regulated gene in OS which is upregulated by the respective drug in the column.

6
R.C. Andrade et al. Computers in Biology and Medicine 134 (2021) 104470

Table 2 Table 2 (continued )


Gene set enrichment analysis results for gene expression profile in the test Description NES FDR Genes
group. The top 20 results are shown, ranked by FDR. The GSEA was performed
using the KEGG database. Pathways that were also enriched in the ORA analysis BECN1, CALCOCO2,
HIF1A, ATG9A, RELA,
(Fig. 2B) are highlighted in bold letters.
MAPK9, RAB7A, ATF4
Description NES FDR Genes Galactose metabolism − 1.8345 0.0067080 PFKP, PGM1, HK1, GLB1,
GAA, UGP2, AKR1B1,
Ashtma 2.5295 0 HLA-DRA, HLA-DRB1,
G6PC3, PFKM, GALT,
HLA-DRB5, HLA-DPA1,
B4GALT2, B4GALT1, PFKL,
FCER1G, HLA-DQA1,
AKR1B10, GALE
HLA-DPB1, HLA-DMB,
p53 signaling ¡1.8398 0.0085862 SERPINE1, IGFBP3,
HLA-DMA, HLA-DQB1
pathway CDKN1A, PERP, THBS1,
Type I diabetes 2.2913 0 HLA-DRA, CPE, HLA-
CCND1, FAZ, ZMAT3,
mellitus DRB1, HLA-DRB5, HLA-
TP53, GADD45A, CCNG1,
DPA1, HLA-DQA1, HLA-
DDB2, BAX, BCL2L1
DPB1, HLA-DMB, HLA-
Nicotinate and − 1.7658 0.017530 NT5E, BST1, AOX1, NNMT,
DMA, HLA-DQB1, CD86
nicotinamide NMRK1
Autoimmune thyroid 2.2776 0 HLA-DRA, HLA-DRB1, HLA-
metabolism
disease DRB5, HLA-DPA1, HLA-
Amino sugar and − 1.7493 0.019012 UGDH, CYB5R3, GFPT2,
DQA1, HLA-DPB1, HLA-
nucleotide sugar PGM1, HK1, PMM1, GMDS,
DMB, HLA-DMA, HLA-
metabolism NAGK, GNPDA1, UGP2,
DQB1, CD86
UAP1L1, HEXB, GFPT1,
Staphylococcus 2.2761 0 HLA-DRA, HLA-DRB1,
GALT, GMPPA, TSTA3,
aureus infection C1QA, HLA-DRB5, HLA-
CYB5R2, UAP1, CYB5R1,
DPA1, FCGR2A, C1QB,
AMDHD2, HEXA, GMPPB,
HLA-DQA1, HLA-DPB1,
GALE, UXS1, PGM3, MPI
C3, FCGR2B, HLA-DMB,
Glycolysis/ − 1.7662 0.021036 BPGM, PFKP, PGM1, HK1,
C3AR1, HLA-DMA, HLA-
Gluconeogenesis ADH5, LDHA, PCK2,
DQB1, FPR3, FCGR1A,
ALDH3A2, ALDH1A3,
C5AR1, ITGB2, ITGAM
ALDH9A1, PKM, ALDH3B1,
Leishmaniasis 2.2696 0 HLA-DRA, HLA-DRB1,
G6PC3, ALDH7A1,
HLA-DRB5, HLA-DPA1,
ALDH1B1, PDHB, ALDOA,
FCGR2A, HLA-DQA1,
ADH1B, PFKM, DLD,
HLA-DPB1, CYBB, ITGA4,
ALDH2, GAPDH, PFKL,
C3, HLA-DMB, HLA-DMA,
PGAM1, ENO1, ALDOC
MARCKSL1, HLA-DQB1,
Steroid biosynthesis − 1.7130 0.031125 DHCR24, MSMO1, LSS,
TGFB3, TLR2, FCGR1A,
DHCR7, CYP51A1, FDFT1,
NCF2, FOS, ITGB2, NCF4,
SOAT1, SC5D
ITGAM, PTPN6
Ascorbate and aldarate − 1.7013 0.033868 UGDH, ALDH3A2,
Allograft rejection 2.2370 0 HLA-DRA, HLA-DRB1,
metabolism ALDH9A1, ALDH7A1,
HLA-DRB5, HLA-DPA1,
ALDH1B1, RGN, ALDH2
HLA-DQA1, HLA-DPB1,
Melanoma ¡1.6734 0.047439 FGF5, FGF7, FGF2,
HLA-DMB, HLA-DMA,
CDKN1A, CCND1, MET,
HLA-DQB1, CD86
TP53, GADD45A, MAPK3,
Systemic lupus 2.1488 0.00010467 HLA-DRA, HLA-DRB1,
DDB2, BAX, EGFR,
erythematosus C1QA, HLA-DRB5, HLA-
MAP2K2, AKT3, HRAS,
DPA1, FCGR2A, C1QB,
MAPK1, PDGFRB,
HLA-DQA1, HLA-DPB1,
PDGFRA
C3, HLA-DMB, HLA-DMA,
HLA-DQB1, FCGR1A, NES: normalized enrichment score.
CD86
Antigen processing and 2.2060 0.00011962 HLA-DRA, CD74, HLA-
presentation DRB1, HLA-DRB5, HLA- is plausible, since the pRb pathway is the most frequently inactivated
DPA1, HLA-DQA1, HLA- pathway in OS, and represents a particularly interesting therapeutic
DPB1, HLA-DMB, CTSS, target [34]. MDFI also binds to the axin complex, resulting in increased
HLA-DMA, HLA-DQB1,
levels of free β-catenin in the cell [33]. High levels of β-catenin were
CD4, LGMN, IFI30, HSPA6,
HSPA2, KLRC4, KLRC2 identified in osteosarcoma tissues compared to normal bone and were
Graft-versus-host 2.0068 0.00075361 HLA-DRA, HLA-DRB1, associated with worse prognosis and lung metastasis [35,36]. High
disease HLA-DRB5, HLA-DPA1, expression of MDFI is associated with unfavorable prognosis in renal
HLA-DQA1, HLA-DPB1, cancer, lung cancer and melanoma [37].
HLA-DMB, HLA-DMA,
HLA-DQB1, CD86
The top down-regulated gene was TPD52L1 (D53), involved in cell
Intestinal immune 2.0141 0.00083735 HLA-DRA, HLA-DRB1, proliferation and calcium signaling. This gene encodes a member of the
network for IgA HLA-DRB5, HLA-DPA1, D52-like family of adaptor proteins that contain a coiled-coil domain,
production CXCR4, HLA-DQA1, HLA- and may form hetero- or homomers. Unlike other D52-like family
DPB1, ITGA4, HLA-DMB,
members, TPD52L1 is not over-expressed in tumor samples [38,39],
HLA-DMA, HLA-DQB1,
CD86 having a more compatible role as a tumor suppressor gene [40]. It also
Steroid hormone − 1.9950 0.0010733 AKR1C3, CYP1B1, interacts with the mitogen-activated protein kinase 5 (MAP3K5/ASK1)
biosynthesis AKR1C2,AKR1C1, STS, and positively regulates MAP3K5-induced apoptosis. One of its isoforms
HSD17B12, COMT activates apoptosis signal-regulating kinase 1 (ASK1), leading to the
Mitophagy − 1.9288 0.0021465 CITED2, RRAS, TP53,
OPTN, SQSTM1, BCL2L1,
activation of the caspase-3-dependent apoptosis pathway [41]. It is
CSNK2A2, TFE3, PINK1, down-regulated in recurrent nasopharyngeal cancer [40], and when
HRAS, RRAS2, NBR1, highly expressed in renal and thyroid cancer, it is associated with
GABARAP, BNIP3L, favorable prognosis [37].
TBC1D17, FIS1, BCL2L13,
Among the commonly reported genes with altered expression in OS,
we found the overexpression of HEY1 (Supplementary Table 1), which is

7
R.C. Andrade et al. Computers in Biology and Medicine 134 (2021) 104470

Table 3
Drugs predicted to reverse at least 10% of OS gene signature and the respective genes with the expression modified by them.
Drug PubChem OS up/drug down OS down/drug up
CID

Afatinib 10184653 ARHGDIB, CD24, CD93, CXCR4, HLA-DPA1, HLA- ADM, AKR1C1, AKR1C2, AKR1C3, ANPEP, CCND1, CDKN1A, CRYAB, CYP1B1, G6PD,
DQA1, LAPTM5, LRRC17, RGS5, SPARCL1, HMOX1, IGFBP3, IGFBP6, ITGBL1, KRT19, LRP10, MFAP5, NQO1, PENK, PTX3, STC2,
TMSB15A TMEM47, UGDH, VAT1
BRD- 3242434 A2M, CD74, HEY1, HLA-DPA1, HLA-DPB1, HLA- ADM, CDKN1A, CHI3L1, COMP, DDAH1, FAS, FHL2, GAS6, GEM, GREM1, HMOX1,
K95196255 DRA, IQGAP2, NES, PTPRZ1, SNX10, TSPAN13 IGFBP3, NT5E, PHLDA3, SERPINE1, STC2, SULF1, TNFRSF12A
DG-041 (DTSI) 11296282 A2M, CD14, CD36, CD93, CXCL14, GMFG, LRRC17, ADAMTS1, ADM, AOX1, BPGM, CAV1, EMP1, FHL2, FST, GEM, PRSS23, SEMA3C,
PECAM1, RGS5, TNFSF10, TYROBP, WIF1 SERPINE1, STC2, THBS1, THBS2, TNFRSF12A
CA-074 methyl 23760717 CA2, CD24, CD74, CXCL14, HEY1, HLA-DPA1, HLA- ADM, AKR1C1, AKR1C2, AKR1C3, C1S, CDKN1A, CHI3L1, FAZ, G6PD, GCLM, HMOX1,
ester DPB1, HLA-DQA1, HLA-DRA, LYZ, RGS5 IGFBP6, IL6, ISLR, NPR3, NQO1, PLIN3, PTX3, SPOCK1, STC2, TM4SF1, UGDH

a downstream effector in the Notch pathway. The Notch pathway afatinib (Gilotrif™) was first approved by the FDA in 2013 and it is still
upregulation has been described in OS, with a possible correlation with currently used as a first line treatment for metastatic non-small-cell lung
invasiveness potential [12]. Other commonly reported genes, like cancer (NSCLC) with EGFR non-resistant activating mutations (most
Runx2, EZR, and HER-2 [12], were not identified as DEGs in our study. commonly exon 19 deletions or exon 21 point mutation L858R) [53,54].
These discrepancies may be due to the enlargement of the sample size in Afatinib has recently demonstrated to decrease proliferation, migration,
our analysis when compared to individual studies, as well as to intra­ and invasion of non-metastatic and metastatic OS cell lines [55], and it is
tumor and intertumor heterogeneity. currently being investigated in a phase II clinical trial for pediatric tu­
The complex microenvironment of OS plays an important role in the mors (https://clinicaltrials.gov/ct2/show/NCT02372006). A different
malignant transformation and maintenance of the tumor. It includes methodology of drug repurposing, using sarcospheres from highly
different types of cells, like bone, vascular and immune cells, which metastatic human OS cell lines and an anticancer drug library, also
communicate through physical contact, soluble factors release or found afatinib as one of the main drugs with therapeutic potential [56].
extracellular vesicles [9,42]. In this study, many of the upregulated Interestingly, the Erb pathway was not enriched in our analysis, and the
pathways were associated to inflammatory response and immunomo­ genes predicted to have its expression altered by afatinib in the samples
dulation. There is a crucial dialogue between bone cells and cells of the evaluated were different from the ones usually reported in the literature.
immune system. Osteosarcomas secrete osteoclastogenic cytokines, like Thus, afatinib may have an inhibitory effect on osteosarcoma cells by
RANKL and macrophage colony-stimulating factor (CSF1) that induce acting in more than one molecular pathway.
bone resorption, while tumor growth is maintained by factors generated The three other top molecules assessed in our analysis are in earlier
during osteolysis [8]. The CSF1 receptor (CSF1R) was upregulated in our phases of study than afatinib. None of them is currently approved by the
analysis, together with the downregulation of TNFRSF11B (Supple­ FDA. BRD-K95196255 is a hydroxyquinoline with in vitro effects that
mentary Tables 1 and 2), which encodes a negative regulator of bone include cytotoxic activity and lung tumor cell growth inhibition [26].
resorption that is inhibited by RANKL. DG-041 is an antagonist of the prostaglandin (PG)E2 EP3 receptor and
One of the modifications that occur in the in microenvironment of OS inhibits platelet aggregation. A potential antimetastatic effect by
is the malignant osteoid formation in the extracellular matrix (ECM), avoiding platelets-induced mesenchymal-like changes in cancer cells has
responsible for the tumor radiographic “sunburst” pattern [42], which is been reported [57]. CA-074me is a methyl ester derivative of CA-074
consistent with DEGs involved in processes such as ECM-receptor with better cell permeability, that has inhibitory activity over the pro­
interaction, as revealed by our ORA analysis. Another important alter­ teases Cathepsin B and L [58]. The degradation of the extracellular
ation is the down-regulation of the p53 pathway, also revealed in our matrix induced by cathepsins seems to contribute to the development
ORA analysis, since the inactivation of TP53 is considered one of the and metastasis of tumors, making them interesting therapeutic targets
major driver events in OS. Germline mutations in TP53 are associated [58–60]. Importantly, CA-074 methyl ester has been shown to inhibit
with the autosomal dominant hereditary cancer condition known as osteoclastogenesis in vitro and in vivo, by mechanisms that seem inde­
Li-Fraumeni syndrome (LFS) that predisposes to a high risk for the pendent from cathepsins, and its use could be advantageous in treating
development of many cancers. Osteosarcoma is one of the core cancers osteolytic conditions, including bone cancers [61].
of LFS, affecting ~16% of the individuals with TP53 germline mutations In conclusion, this study brings new insights into the biological
[43]. Besides, somatic rearrangements of intron 1 of TP53 are thought to knowledge of osteosarcoma, an aggressive cancer with limited thera­
be a specific genetic alteration for OS, occurring in ~16% of the sporadic peutic options. We were able to identify a gene expression signature for
OS samples [44]. this tumor using data from a large number of tumor samples from young
We assessed possible new candidate drugs for the treatment of OS individuals and comparing them to both candidate cells of origin: os­
based on the gene signature obtained in our meta-analysis. For that teoblasts and mesenchymal stem cells. We were then able to propose
intent, we used the LINCS CDS2 repurposing tool, which is considered an new therapeutic small molecules based on this gene expression signa­
updated and curated tool for drug repurposing based on gene expression ture. Clearly, drug repurposing based solely on bioinformatics approach
signature [24,45–48]. For further discussion, we selected the top four has limitations, since the efficacy of a drug depends on several variables
drug candidates obtained using the LINCS CDS2 engine, which were other than gene expression signature, and further in vitro and in vivo
predicted to reverse the expression of at least 10% of our input genes (i. studies are necessary to establish the potential of these drugs in OS
e.: with a drug score higher than 10%). There is not a threshold deter­ treatment. However, given the modest improvement in OS treatment
mined in terms of significance for this score [24]. The users of the LINCS over the last decades, we believe our results can be an important
CDS2 engine can select, for further discussion/filtering, all the 50 drugs contribution for the investigation of new therapeutic genetic targets and
in the output, the top drugs among them, or even the ones that have for selecting new drugs to be tested for this disease.
more published information available, independently of the score
[49–52]. The best ranked drug in our study was afatinib (S1011; Pub­ Declaration of competing interest
Chem CID 10184653), a selective tyrosine kinase inhibitor that cova­
lently binds to and irreversibly blocks signaling from the ErbB family The authors declare no conflicts of interest.
members EGFR (ErbB1), HER2 (ErbB2) and HER4 (ErbB4) [53,54]. Oral

8
R.C. Andrade et al. Computers in Biology and Medicine 134 (2021) 104470

Acknowledgments [14] B.S. Carvalho, R.A. Irizarry, A framework for oligonucleotide microarray
preprocessing, Bioinformatics 26 (2010) 2363–2367, https://doi.org/10.1093/
bioinformatics/btq431.
We thank Nicole de Miranda Scherer, Daniel Andrade Moreira and [15] T.L. Leek Jt, W.E. Johnson, H.S. Parker, E.J. Fertig, A.E. Jaffe, Y. Zhang, J.
Jéssica Gonçalves Vieira da Cruz for the orientations during the D. Storey, Sva: Surrogate Variable Analysis, in: R Package, 2020, version 3.36.0.
computational analyses. We also thank Inova Fiocruz for the financial [16] M.E. Ritchie, B. Phipson, D. Wu, Y. Hu, C.W. Law, W. Shi, G.K. Smyth, Limma
powers differential expression analyses for RNA-sequencing and microarray
support. This work is dedicated to the memory of Nilton Barreto dos studies, Nucleic Acids Res. 43 (2015) e47, https://doi.org/10.1093/nar/gkv007.
Santos, dear friend and great scientist. [17] E. Karatzas, G. Kolios, G.M. Spyrou, An Application of Computational Drug
Repurposing Based on Transcriptomic Signatures, in: Methods Mol. Biol., Humana
Press Inc., 2019, pp. 149–177, https://doi.org/10.1007/978-1-4939-8955-3_9.
Appendix A. Supplementary data [18] W. Yang, Y. Li, F. He, H. Wu, Microarray profiling of long non-coding RNA
(lncRNA) associated with hypertrophic cardiomyopathy, BMC Cardiovasc. Disord.
Supplementary data to this article can be found online at https://doi. 15 (2015) 1–9, https://doi.org/10.1186/s12872-015-0056-7.
[19] P. Wang, Y. Wang, B. Hang, X. Zou, J.H. Mao, A novel gene expression-based
org/10.1016/j.compbiomed.2021.104470. prognostic scoring system to predict survival in gastric cancer, Oncotarget 7 (2016)
55343–55351, https://doi.org/10.18632/oncotarget.10533.
Authors contributions [20] K. Zhang, H. Shi, H. Xi, X. Wu, J. Cui, Y. Gao, W. Liang, C. Hu, Y. Liu, J. Li,
N. Wang, B. Wei, L. Chen, Genome-wide lncRNA microarray profiling identifies
novel circulating lncrnas for detection of gastric cancer, Theranostics 7 (2017)
Conception and design: RCA, MB and FRV; Data survey and selec­ 213–227, https://doi.org/10.7150/thno.16044.
tion: RCA and MKA; Analysis and interpretation: RCA and MB; Drafting [21] S. Zhang, X. Zeng, T. Ding, L. Guo, Y. Li, S. Ou, H. Yuan, Microarray profile of
circular RNAs identifies hsa-circ-0014130 as a new circular RNA biomarker in non-
of the article: RCA; Review and editing of the article: MB, MKA and FRV.
small cell lung cancer, Sci. Rep. 8 (2018) 2878, https://doi.org/10.1038/s41598-
018-21300-5.
Funding source [22] Y.J. Zhu, B. Zheng, G.J. Luo, X.K. Ma, X.Y. Lu, X.M. Lin, S. Yang, Q. Zhao, T. Wu, Z.
X. Li, X.L. Liu, R. Wu, J.F. Liu, Y. Ge, L. Yang, H.Y. Wang, L. Chen, Circular RNAs
negatively regulate cancer stem cells by physically binding FMRP against CCAR1
Inova Fiocruz/Fundação Oswaldo Cruz, Rio de Janeiro, Brazil (Ju­ complex in hepatocellular carcinoma, Theranostics 9 (2019) 3526–3540, https://
nior Postdoctoral fellowship 2019-2020). The funding source was not doi.org/10.7150/thno.32796.
involved in the study design, data analysis and preparation of the article. [23] Y. Liao, J. Wang, E.J. Jaehnig, Z. Shi, B. Zhang, WebGestalt, Gene set analysis
toolkit with revamped UIs and APIs, Nucleic Acids Res. 47 (2019) W199–W205,
https://doi.org/10.1093/nar/gkz401, 2019.
References [24] Q. Duan, S.P. Reid, N.R. Clark, Z. Wang, N.F. Fernandez, A.D. Rouillard,
B. Readhead, S.R. Tritsch, R. Hodos, M. Hafner, M. Niepel, P.K. Sorger, J.T. Dudley,
[1] D.M. Gianferante, L. Mirabello, S.A. Savage, Germline and somatic genetics of S. Bavari, R.G. Panchal, A. Ma’Ayan, LINCS L1000 characteristic direction
osteosarcoma - connecting aetiology, biology and therapy, Nat. Rev. Endocrinol. 13 signatures search engine, L1000CDS2, Npj Syst. Biol. Appl. 2 (2016), https://doi.
(2017) 480–491, https://doi.org/10.1038/nrendo.2017.16. org/10.1038/npjsba.2016.15.
[2] A. Misaghi, A. Goldin, M. Awad, A.A. Kulidjian, Osteosarcoma: a comprehensive [25] NIH, LINCS data portal | cells - L1000 mRNA profiling assay. http://lincsportal.ccs.
review, SICOT-J. 4 (2018), https://doi.org/10.1051/sicotj/2017028. miami.edu/cells/#/catalog?query=assays:L1000 mRNA profiling assay (accessed
[3] S. Chen, L. Yang, F. Pu, H. Lin, B. Wang, J. Liu, Z. Shao, High birth weight increases April 18, 2021).
the risk for bone tumor: a systematic review and meta-analysis, Int. J. Environ. Res. [26] S. Kim, J. Chen, T. Cheng, A. Gindulyte, J. He, S. He, Q. Li, B.A. Shoemaker, P.
Publ. Health 12 (2015) 11178–11195, https://doi.org/10.3390/ijerph120911178. A. Thiessen, B. Yu, L. Zaslavsky, J. Zhang, E.E. Bolton, PubChem 2019 update:
[4] S. Prater, B. McKeon, Cancer, Osteosarcoma, StatPearls Publishing, 2019. http improved access to chemical data, Nucleic Acids Res. 47 (2019) D1102–D1109,
://www.ncbi.nlm.nih.gov/pubmed/31751058. (Accessed 2 July 2020). https://doi.org/10.1093/nar/gky1033.
[5] M.S. Isakoff, S.S. Bielack, P. Meltzer, R. Gorlick, Osteosarcoma: current treatment [27] E. Simpson, H.L. Brown, Understanding osteosarcomas, JAAPA 31 (2018) 15–19,
and a collaborative pathway to success, J. Clin. Oncol. 33 (2015) 3029–3035, https://doi.org/10.1097/01.JAA.0000541477.24116.8d.
https://doi.org/10.1200/JCO.2014.59.4895. [28] K.M. Makielski, L.J. Mills, A.L. Sarver, M.S. Henson, L.G. Spector, S. Naik, J.
[6] S. Smeland, S.S. Bielack, J. Whelan, M. Bernstein, P. Hogendoorn, M.D. Krailo, F. Modiano, Risk factors for development of canine and human osteosarcoma: a
R. Gorlick, K.A. Janeway, F.C. Ingleby, J. Anninga, I. Antal, C. Arndt, K.L.B. Brown, comparative review, Vet. Sci. 6 (2019), https://doi.org/10.3390/vetsci6020048.
T. Butterfass-Bahloul, G. Calaminus, M. Capra, C. Dhooge, M. Eriksson, A. [29] D.C. Stefan, M. Harif, Pediatr. Cancer Africa, Osteosarcoma, Springer International
M. Flanagan, G. Friedel, M.C. Gebhardt, H. Gelderblom, R. Goldsby, H.E. Grier, Publishing, Cham, 2017, pp. 71–80, https://doi.org/10.1007/978-3-319-17936-0_
R. Grimer, D.S. Hawkins, S. Hecker-Nolting, K. Sundby Hall, M.S. Isakoff, G. Jovic, 6.
T. Kühne, L. Kager, T. von Kalle, E. Kabickova, S. Lang, C.C. Lau, P.J. Leavey, S. [30] B. Otoukesh, B. Boddouhi, M. Moghtadaei, P. Kaghazian, M. Kaghazian, Novel
L. Lessnick, L. Mascarenhas, R. Mayer-Steinacker, P.A. Meyers, R. Nagarajan, R. molecular insights and new therapeutic strategies in osteosarcoma 11 Medical and
L. Randall, P. Reichardt, M. Renard, C. Rechnitzer, C.L. Schwartz, S. Strauss, Health Sciences 1112 Oncology and Carcinogenesis, Canc. Cell Int. 18 (2018) 1–23,
L. Teot, B. Timmermann, M.R. Sydes, N. Marina, Survival and prognosis with https://doi.org/10.1186/s12935-018-0654-4.
osteosarcoma: outcomes in more than 2000 patients in the EURAMOS-1 (European [31] A. Ertel, A. Verghese, S.W. Byers, M. Ochs, A. Tozeren, Pathway-specific
and American Osteosarcoma Study) cohort, Eur. J. Canc. 109 (2019) 36–50, differences between tumor cell lines and normal and tumor tissue cells, Mol. Canc.
https://doi.org/10.1016/j.ejca.2018.11.027. 5 (2006) 55, https://doi.org/10.1186/1476-4598-5-55.
[7] R.D. Roberts, M.M. Lizardo, D.R. Reed, P. Hingorani, J. Glover, W. Allen-Rhoades, [32] A. Longhi, C. Errani, D. Gonzales-Arabio, C. Ferrari, M. Mercuri, Osteosarcoma in
T. Fan, C. Khanna, E.A. Sweet-Cordero, T. Cash, M.W. Bishop, M. Hegde, A. patients older than 65 years, J. Clin. Oncol. 26 (2008) 5368–5373, https://doi.org/
R. Sertil, C. Koelsche, L. Mirabello, D. Malkin, P.H. Sorensen, P.S. Meltzer, K. 10.1200/JCO.2007.14.9104.
A. Janeway, R. Gorlick, B.D. Crompton, Provocative questions in osteosarcoma [33] A. Bateman, UniProt: a worldwide hub of protein knowledge, Nucleic Acids Res. 47
basic and translational biology: a report from the Children’s Oncology Group, (2019) D506–D515, https://doi.org/10.1093/nar/gky1049.
Cancer 125 (2019) 3514–3525, https://doi.org/10.1002/cncr.32351. [34] S.E. Ballatori, P.W. Hinds, Osteosarcoma: prognosis plateau warrants
[8] M. Kansara, M.W. Teng, M.J. Smyth, D.M. Thomas, Translational biology of retinoblastoma pathway targeted therapy, Signal Transduct. Target. Ther. 1 (2016)
osteosarcoma, Nat. Rev. Canc. 14 (2014) 722–735, https://doi.org/10.1038/ 16001, https://doi.org/10.1038/sigtrans.2016.1.
nrc3838. [35] W. Liu, Z. Zhao, Y. Wang, W. Li, Q. Su, Q. Jia, J. Zhang, X. Zhang, J. Yin, J. Shen,
[9] A. Abarrategi, J. Tornin, L. Martinez-Cruzado, A. Hamilton, E. Martinez-Campos, J. Dioscin inhibits stem-cell-like properties and tumor growth of osteosarcoma
P. Rodrigo, M.V. González, N. Baldini, J. Garcia-Castro, R. Rodriguez, through Akt/GSK3/β-catenin signaling pathway, Cell Death Dis. 9 (2018), https://
Osteosarcoma: cells-of-origin, cancer stem cells, and targeted therapies, Stem Cell. doi.org/10.1038/s41419-018-0363-x.
Int. (2016), https://doi.org/10.1155/2016/3631764 (2016) 3631764. [36] Y. Lu, G.F. Guan, J. Chen, B. Hu, C. Sun, Q. Ma, Y.H. Wen, X.C. Qiu, Y. Zhou,
[10] A.J. Mutsaers, C.R. Walkley, Cells of origin in osteosarcoma: mesenchymal stem Aberrant CXCR4 and β-catenin expression in osteosarcoma correlates with patient
cells or osteoblast committed cells? Bone 62 (2014) 56–63, https://doi.org/ survival, Oncol. Lett. 10 (2015) 2123–2129, https://doi.org/10.3892/
10.1016/j.bone.2014.02.003. ol.2015.3535.
[11] A.L. Sinatro, Osteosarcoma: Molecular Etiology, Pathophysiology, Clinical [37] M. Uhlén, L. Fagerberg, B.M. Hallström, C. Lindskog, P. Oksvold, A. Mardinoglu,
Presentation, Diagnosis, and Treatment, Boston University, 2018. Å. Sivertsson, C. Kampf, E. Sjöstedt, A. Asplund, I.M. Olsson, K. Edlund,
[12] E. Tirtei, M. Cereda, E. De Luna, P. Quarello, S.D. Asaftei, F. Fagioli, Omic E. Lundberg, S. Navani, C.A.K. Szigyarto, J. Odeberg, D. Djureinovic, J.O. Takanen,
approaches to pediatric bone sarcomas, Pediatr. Blood Canc. 67 (2020), https:// S. Hober, T. Alm, P.H. Edqvist, H. Berling, H. Tegel, J. Mulder, J. Rockberg,
doi.org/10.1002/pbc.28072. P. Nilsson, J.M. Schwenk, M. Hamsten, K. Von Feilitzen, M. Forsberg, L. Persson,
[13] L. Gautier, L. Cope, B.M. Bolstad, R.A. Irizarry, Affy - analysis of Affymetrix F. Johansson, M. Zwahlen, G. Von Heijne, J. Nielsen, F. Pontén, Tissue-based map
GeneChip data at the probe level, Bioinformatics 20 (2004) 307–315, https://doi. of the human proteome, Science 80– (2015) 347, https://doi.org/10.1126/
org/10.1093/bioinformatics/btg405. science.1260419.
[38] M. Shehata, I. Bièche, R. Boutros, J. Weidenhofer, S. Fanayan, L. Spalding, N. Zeps,
K. Byth, R.K. Bright, R. Lidereau, J.A. Byrne, Nonredundant functions for tumor

9
R.C. Andrade et al. Computers in Biology and Medicine 134 (2021) 104470

protein D52-like proteins support specific targeting of TPD52, Clin. Canc. Res. 14 [48] O.S. Kwon, W. Kim, H.J. Cha, H. Lee, Silico drug repositioning: from large-scale
(2008) 5050–5060, https://doi.org/10.1158/1078-0432.CCR-07-4994. transcriptome data to therapeutics, Arch Pharm. Res. (Seoul) 42 (2019) 879–889,
[39] D. Barbaric, K. Byth, L. Dalla-Pozza, J.A. Byrne, Expression of tumor protein D52- https://doi.org/10.1007/s12272-019-01176-3.
like genes in childhood leukemia at diagnosis: clinical and sample considerations, [49] P. Fagone, R. Caltabiano, A. Russo, G. Lupo, C.D. Anfuso, M.S. Basile, A. Longo,
Leuk. Res. 30 (2006) 1355–1363, https://doi.org/10.1016/j.leukres.2006.03.009. F. Nicoletti, R. De Pasquale, M. Libra, M. Reibaldi, Identification of novel
[40] Z. Huang, W. Li, S. Lin, X. Fang, C. Zhang, Z. Liao, Identification of novel tumor chemotherapeutic strategies for metastatic uveal melanoma, Sci. Rep. 7 (2017),
suppressor genes down-regulated in recurrent nasopharyngeal cancer by DNA https://doi.org/10.1038/srep44564.
microarray, Indian J. Otolaryngol. Head Neck Surg. 66 (2014) 120–125, https:// [50] Z. Wang, A. Ma’ayan, An open RNA-Seq data analysis pipeline tutorial with an
doi.org/10.1007/s12070-011-0359-7. example of reprocessing data from a recent Zika virus study [version 1; referees: 3
[41] S. Cho, H.M. Ko, J.M. Kim, J.A. Lee, J.E. Park, M.S. Jang, S.G. Park, D.H. Lee, S. approved, F1000Research (5) (2016), https://doi.org/10.12688/
E. Ryu, B.C. Park, Positive regulation of apoptosis signal-regulating kinase 1 by F1000RESEARCH.9110.1.
hD53L1, J. Biol. Chem. 279 (2004) 16050–16056, https://doi.org/10.1074/jbc. [51] A.A. Pezzulo, R.A. Tudas, C.G. Stewart, L.G. Vargas Buonfiglio, B.D. Lindsay, P.
M305758200. J. Taft, N.D. Gansemer, J. Zabner, HSP90 inhibitor geldanamycin reverts IL-13–and
[42] H.K. Brown, K. Schiavone, F. Gouin, M.F. Heymann, D. Heymann, Biology of bone IL-17–induced airway goblet cell metaplasia, J. Clin. Invest. 129 (2019) 744–758,
sarcomas and new therapeutic developments, Calcif. Tissue Int. 102 (2018) https://doi.org/10.1172/JCI123524.
174–195, https://doi.org/10.1007/s00223-017-0372-2. [52] B. Turanli, K. Karagoz, G. Bidkhori, R. Sinha, M.L. Gatza, M. Uhlen, A. Mardinoglu,
[43] G. Bougeard, M. Renaux-Petel, J.-M. Flaman, C. Charbonnier, P. Fermey, K.Y. Arga, Multi-omic data interpretation to repurpose subtype specific drug
M. Belotti, M. Gauthier-Villars, D. Stoppa-Lyonnet, E. Consolino, L. Brugières, candidates for breast cancer, Front. Genet. 10 (2019) 420, https://doi.org/
O. Caron, P.R. Benusiglio, B. Bressac-de Paillerets, V. Bonadona, C. Bonaïti-Pellié, 10.3389/fgene.2019.00420.
J. Tinat, S. Baert-Desurmont, T. Frebourg, Revisiting Li-fraumeni syndrome from [53] R.T. Dungo, G.M. Keating, Afatinib: first global approval, Drugs 73 (2013)
TP53 mutation carriers, J. Clin. Oncol. 33 (2015) 2345–2352, https://doi.org/ 1503–1515, https://doi.org/10.1007/s40265-013-0111-6.
10.1200/JCO.2014.59.5728. [54] D.S. Wishart, Y.D. Feunang, A.C. Guo, E.J. Lo, A. Marcu, J.R. Grant, T. Sajed,
[44] S. Ribi, D. Baumhoer, K. Lee, Edison, A.S.M. Teo, B. Madan, K. Zhang, W. D. Johnson, C. Li, Z. Sayeeda, N. Assempour, I. Iynkkaran, Y. Liu, A. MacIejewski,
K. Kohlmann, F. Yao, W.H. Lee, Q. Hoi, S. Cai, X.Y. Woo, P. Tan, G. Jundt, J. Smida, N. Gale, A. Wilson, L. Chin, R. Cummings, Di Le, A. Pon, C. Knox, M. Wilson,
M. Nathrath, W.K. Sung, J.D. Schiffman, D.M. Virshup, A.M. Hillmer, TP53 intron 1 DrugBank 5.0: a major update to the DrugBank database for 2018, Nucleic Acids
hotspot rearrangements are specific to sporadic osteosarcoma and can cause Li- Res. 46 (2018) D1074–D1082, https://doi.org/10.1093/nar/gkx1037.
Fraumeni syndrome, Oncotarget 6 (2015) 7727–7740, https://doi.org/10.18632/ [55] M. Cruz-Ramos, Y. Zamudio-Cuevas, D. Medina-Luna, K. Martínez-Flores,
oncotarget.3115. G. Martínez-Nava, J. Fernández-Torres, A. López-Reyes, F. Solca, Afatinib is active
[45] A.B. Keenan, S.L. Jenkins, K.M. Jagodnik, S. Koplev, E. He, D. Torre, Z. Wang, A. in osteosarcoma in osteosarcoma cell lines, J. Canc. Res. Clin. Oncol. 146 (2020)
B. Dohlman, M.C. Silverstein, A. Lachmann, M.V. Kuleshov, A. Ma’ayan, 1693–1700, https://doi.org/10.1007/s00432-020-03220-y.
V. Stathias, R. Terryn, D. Cooper, M. Forlin, A. Koleti, D. Vidovic, C. Chung, S. [56] Annual meeting of the orthopaedic research society orlando, FL march 5-8, 2016,
C. Schürer, J. Vasiliauskas, M. Pilarczyk, B. Shamsaei, M. Fazel, Y. Ren, W. Niu, N. 2016, J. Orthop. Res. 34 (2016) S1, https://doi.org/10.1002/jor.23247. S1
A. Clark, S. White, N. Mahi, L. Zhang, M. Kouril, J.F. Reichard, S. Sivaganesan, (abstract 0384, by Collier CD et al).
M. Medvedovic, J. Meller, R.J. Koch, M.R. Birtwistle, R. Iyengar, E.A. Sobie, E. [57] P. Guillem-Llobat, M. Dovizio, A. Bruno, E. Ricciotti, V. Cufino, A. Sacco,
U. Azeloglu, J. Kaye, J. Osterloh, K. Haston, J. Kalra, S. Finkbiener, J. Li, P. Milani, R. Grande, S. Alberti, V. Arena, M. Cirillo, C. Patrono, G.A. FitzGerald,
M. Adam, R. Escalante-Chong, K. Sachs, A. Lenail, D. Ramamoorthy, E. Fraenkel, D. Steinhilber, A. Sgambato, P. Patrignani, Aspirin prevents colorectal cancer
G. Daigle, U. Hussain, A. Coye, J. Rothstein, D. Sareen, L. Ornelas, M. Banuelos, metastasis in mice by splitting the crosstalk between platelets and tumor cells,
B. Mandefro, R. Ho, C.N. Svendsen, R.G. Lim, J. Stocksdale, M.S. Casale, T. Oncotarget 7 (2016) 32462–32477, https://doi.org/10.18632/oncotarget.8655.
G. Thompson, J. Wu, L.M. Thompson, V. Dardov, V. Venkatraman, A. Matlock, J. [58] H. Ruan, S. Hao, P. Young, H. Zhang, Targeting Cathepsin B for Cancer Therapies,
E. Van Eyk, J.D. Jaffe, M. Papanastasiou, A. Subramanian, T.R. Golub, S. in: Horizons Cancer Res., Nova Science Publishers, Inc., 2015, pp. 23–40. PMID:
D. Erickson, M. Fallahi-Sichani, M. Hafner, N.S. Gray, J.R. Lin, C.E. Mills, J. 26623174.
L. Muhlich, M. Niepel, C.E. Shamu, E.H. Williams, D. Wrobel, P.K. Sorger, L. [59] D.R. Sudhan, D.W. Siemann, Cathepsin L targeting in cancer treatment, Pharmacol.
M. Heiser, J.W. Gray, J.E. Korkola, G.B. Mills, M. LaBarge, H.S. Feiler, M.A. Dane, Ther. 155 (2015) 105–116, https://doi.org/10.1016/j.pharmthera.2015.08.007.
E. Bucher, M. Nederlof, D. Sudar, S. Gross, D.F. Kilburn, R. Smith, K. Devlin, [60] D.R. Sudhan, C. Pampo, L. Rice, D.W. Siemann, Cathepsin L inactivation leads to
R. Margolis, L. Derr, A. Lee, A. Pillai, The library of integrated network-based multimodal inhibition of prostate cancer cell dissemination in a preclinical bone
cellular signatures NIH program: system-level cataloging of human cells response metastasis model, Int. J. Canc. 138 (2016) 2665–2677, https://doi.org/10.1002/
to perturbations, Cell Syst 6 (2018) 13–24, https://doi.org/10.1016/j. ijc.29992.
cels.2017.11.001. [61] N. Patel, S. Nizami, L. Song, M. Mikami, A. Hsu, T. Hickernell,
[46] A. Musa, L.S. Ghoraie, S.-D. Zhang, G. Galzko, O. Yli-Harja, M. Dehmer, B. Haibe- C. Chandhanayingyong, S. Rho, J.T. Compton, J.M. Caldwell, P.B. Kaiser, H. Bai, H.
Kains, F. Emmert-Streib, A review of connectivity map and computational G. Lee, C.R. Fischer, F.Y. Lee, CA-074Me compound inhibits osteoclastogenesis via
approaches in pharmacogenomics, Briefings Bioinf. 19 (2017), https://doi.org/ suppression of the NFATc1 and c-FOS signaling pathways, J. Orthop. Res. 33
10.1093/bib/bbw112 bbw112. (2015) 1474–1486, https://doi.org/10.1002/jor.22795.
[47] K.M. Kingsmore, A.C. Grammer, P.E. Lipsky, Drug repurposing to improve
treatment of rheumatic autoimmune inflammatory diseases, Nat. Rev. Rheumatol.
16 (2020) 32–52, https://doi.org/10.1038/s41584-019-0337-0.

10

You might also like