Open Access

Genetic profiling of osteosarcoma in an adolescent using a next‑generation sequencing panel and Sanger sequencing: A case report and review of the literature

  • Authors:
    • Mariana Chantre‑Justino
    • Rafaele Tavares Silvestre
    • Thiago Luz De Castro
    • Eliane Luz
    • Rafael De Castro E Silva Pinheiro
    • Anabela Caruso
    • Ana Cristina De Sá Lopes
    • Walter Meohas
    • Gilda Alves
    • Maria Helena Faria Ornellas
  • View Affiliations

  • Published online on: January 3, 2025     https://doi.org/10.3892/br.2025.1920
  • Article Number: 42
  • Copyright: © Chantre‑Justino 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

Osteosarcoma (OS) is the most common malignant bone tumor affecting adolescents and young adults and it usually occurs in the long bones of the extremities. The detection of cancer‑related genetic alterations has a growing effect in guiding diagnosis, prognosis and targeted therapies. However, little is known about the molecular aspects involved in the etiology and progression of OS, which limits options for targeted therapies. The present study described a case of an adolescent patient (16‑years‑old) who was diagnosed with conventional central OS in the right distal femur without the evidence of pulmonary metastases; the patient was treated with surgery and adjuvant chemotherapy. Genetic alterations in resected tumor tissue were investigated via next‑generation sequencing (NGS) technology using a targeted NGS panel. Sanger sequencing was also performed to investigate somatic and germline TP53 mutations (exons 4‑8). NGS analysis revealed an intratumor heterogeneity signature in OS tumor, including several single nucleotide variants identified in genes encoding tyrosine kinase proteins. No PCR products for TP53 exon 5 were detected in the tumor sample by PCR analysis prior to Sanger sequencing, suggesting a significant deletion in this exon. Sanger sequencing analysis revealed the missense variant TP53 c.712T>A (p.Cys238Ser) in tumor tissue sample, thus reinforcing the role of TP53 somatic mutations in OS development. Additionally, the TP53 c.215C>G (p.Pro72Arg) germline missense variant was identified in the peripheral blood sample. In conclusion, the findings provided new information on genetic aspects that may contribute to OS development, especially in pediatric patients.

Introduction

Osteosarcoma (OS) is the most common primary malignant bone tumor affecting adolescents and young adults and it predominantly occurs in the long bones of the extremities, notably in the femur (1,2). The lungs are the most common metastatic site for OS and indicate a poor prognosis. OS incidence is rare, accounting for <1% of all human cancers and ~2% of childhood and adolescent cancers (3). OS clinical evaluation includes medical history and physical examination followed by radiologic imaging and tissue biopsy for histological diagnosis (4). The treatment modality generally includes surgery and combination of chemotherapy (5). However, some patients do not respond effectively to therapy; therefore, new treatment modalities are needed. Several other oncological treatments showing promising results in a number of cancers are being investigated for sarcomas, including the use of poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi), new adjuvant therapies, immunotherapy with immune checkpoint inhibitors (blockage of the programmed cell death protein 1/programmed death-ligand 1 axis) and epigenetic therapies (6-14). Despite these efforts, the efficacy or clinical benefits of these treatment strategies for sarcomas remain controversial.

Advances in understanding cancer-related genetic alterations have had substantial effects on precision oncology (15,16). Next-generation sequencing (NGS) is a high-throughput sequencing offering robust genomic data for tumor genotyping that is able to drive diagnosis and treatment decisions (16,17). Despite this, little is known about the molecular aspects involved in OS etiology and progression; thus, genomic testing and targeted therapy to improve treatment are still rare scenarios for patients with OS. Mutations in the TP53 and RB1 tumor suppressor genes are commonly described for OS (18,19). However, the clinical complexity of OS suggests additional genetic drivers of this neoplasm.

The present study described the clinical course and the genomic profiling of an adolescent patient who was diagnosed with OS and treated with a standard protocol. It was approved by the Ethics and Research Committee of the National Institute of Traumatology and Orthopedics (approval no. CAAE: 60632822.4.0000.5273).

Case presentation

A 16-year-old female was referred to the Specialized Care Center for Orthopedic Oncology (National Institute of Traumatology and Orthopedics, Rio de Janeiro, Brazil) in November 2022 due to a mass, swelling and increasing pain in the right knee which began two months earlier. Patient had no known family history of cancer. The physical exam revealed a palpable mass in the right knee and restricted range of motion (10-90˚) associated with pain, as major findings. Radiography revealed a blastic lesion affecting the metadiaphyseal region of the right distal femur with a sunburst periosteal reaction (Fig. 1A). Magnetic resonance imaging revealed infiltration of the lesion through the distal third of the femur and mild involvement of soft tissues, resulting in a lesion that appeared hypointense on T1 and hyperintense on T2 (Fig. 1B). Computed tomography of the chest did not reveal evidence of distant metastases (Fig. 1C). A needle biopsy was performed and the diagnosis of conventional central OS, grade III, with a chondroblastic area was confirmed by histological examination (Fig. 2A and B). Tissue preparation for histological examination was performed by hematoxylin and eosin (H&E) staining. Briefly, the deparaffinization procedure of the tissue section was performed with xylene followed by alcohol rinses and then by rinsing in tap water to hydrate the section. Next, the Harris hematoxylin solution was applied for 2 min to stain the nuclear elements, followed by rinsing in tap water. After, a treatment with 5% acid alcohol was carried out followed by rinsing in tap water and subsequent addition of a 70% alcohol solution for 1 min. Finally, the eosin solution was applied for 15 sec to stain nonnuclear elements, followed by 100% ethanol rinses for dehydration and xylene treatment. For microscopy images, the stained tissues were cut into 7-µm slices using a microtome.

In January 2023, the patient started treatment with a neoadjuvant chemotherapy regimen of six cycles of cisplatin 60 mg/m2, doxorubicin 75 mg/m2, cardioxane 375 mg/m2 and high doses of methotrexate (12 g/m2). In May 2023, the patient underwent a wide resection and endoprosthetic reconstruction (Fig. 1D). Histopathological analysis of the surgical specimen revealed tumor necrosis of 60% (Huvos grade II), tumor-free resection margin and absence of angiolymphatic or perineural invasion (Fig. 2C and D). In June 2023, the patient started adjuvant chemotherapy with twelve cycles of cisplatin 60 mg/m2, doxorubicin 75 mg/m2, cardioxane 375 mg/m2 and high doses of methotrexate (12 g/m2). Until the last follow-up in April 2024, the patient still had good clinical signs, with no evidence of recurrence or pulmonary metastases.

The tumor tissue sample for DNA sequencing analysis was obtained from the surgical resection, 4 months after neoadjuvant chemotherapy. A peripheral blood sample was obtained at the same time. NGS analysis of tumor tissue was performed using the AmpliSeq for Illumina Focus Panel (Illumina, Inc.). Briefly, tumor tissue was fragmented using the L-Beader 24 tissue disruptor (Loccus do Brasil Ltda). Then, genomic DNA was extracted using Quick-DNA Miniprep Kit (Zymo Research Corp.). For NGS, the library preparation process was performed with 10 ng of DNA input, in which DNA targets were amplified by PCR using the AmpliSeq Focus DNA Panel and the AmpliSeq for Illumina Library PLUS (cat. no. 20019101; Illumina, Inc.). The AmpliSeq Focus DNA Panel contains 29 kb and the DNA amplicon size contains an average size of 107 bp in length. Afterwards, the amplicons were partially digested and subsequently ligated with index combination for dual-index sequencing using the AmpliSeq for Illumina CD Indexes Set A (cat. no. 20019105; Illumina, Inc.). All DNA library preparation assay was performed according to the manufacturer's protocol, available on the Illumina website (https://support.illumina.com/downloads/ampliseq-for-illumina-focus-panel-reference-guide-1000000039456.html). The quality of DNA library was visualized on 2% agarose gel electrophoresis using a DNA ladder 100 bp (Ludwig Biotecnologia ltda) and stained with ethidium bromide, revealing a size distribution corresponding to ~300 bp. The quantification of DNA library was assessed by Qubit dsDNA HS Assay Kit (Thermo Fisher Scientific, Inc.). Finally, after denaturing and diluting steps, the library was loaded onto the reagent cartridge and transferred automatically to a flow cell (NextSeq 500/550 Mid-Output v2.5 Kit; cat. no. 20024905; Illumina, Inc.) for paired-end sequencing reads based on sequencing by synthesis (SBS) technology on the NextSeq 550 Sequencing System. For bioinformatics analysis, genomics data were analyzed using the DNA Amplicon App (Version 2.1.1) on BaseSpace Sequence Hub (Illumina, Inc.) (https://www.illumina.com/products/by-type/informatics-products/basespace-sequence-hub/apps/dna-amplicon.html), resulting in genomic variants identified in the following databases: Single Nucleotide Polymorphism Database (dbSNP) (https://www.ncbi.nlm.nih.gov/snp/?cmd=search), Catalogue of Somatic Mutations in Cancer (COSMIC) (https://cancer.sanger.ac.uk/cosmic) and ClinVar (https://www.ncbi.nlm.nih.gov/clinvar/). In the present study, the single nucleotide variants (SNVs) in tumor tissue were identified in coding and intronic regions described in the following genes: JAK1, ALK, FGFR3, PDGFRA, FGFR4, EGFR, RET and KRAS.

The AmpliSeq for Illumina Focus Panel (Illumina, Inc.) includes genes with known relevance to solid tumors. However, the TP53 gene is not part of this panel. Since TP53 mutations are commonly described for OS, mutational hotspot exons of TP53 (exons 4-8) were investigated in the present study by Sanger sequencing. To investigate somatic and germline TP53 mutations, DNA samples were obtained from tumor and peripheral blood. Briefly, genomic DNA was extracted from blood using QIAamp Blood Mini Kit (cat. no. 51104; Qiagen GmbH). The PCR products of TP53 (exons 4-8) were purified by using the PureLink Quick Gel Extraction and PCR Purification Combo Kit (cat. no. K220001; Invitrogen; Thermo Fisher Scientific, Inc.), followed by Sanger sequencing reaction using the BigDye Terminator v3.1 Cycle Sequencing kit on an ABI 3730XL DNA Analyzer (Applied Biosystems; Thermo Fisher Scientific, Inc.), as previously described (20). DNA sequencing results were analyzed with ChromasPro software (Technelysium Pty Ltd), version 2.1.10.1, using the reference sequence from NCBI (NM_000546.6).

As shown in Fig. 3, the PCR products of TP53 were initially visualized on polyacrylamide gels. Only TP53 exon 5 was not amplified from the tumor tissue sample, which suggested a significant somatic exonic deletion of TP53 in this OS tumor. Sanger sequencing analysis revealed the missense variant TP53 c.712T>A (p.Cys238Ser) in the tumor sample (Fig. 4) and the missense variant TP53 c.215C>G (p.Pro72Arg) in the peripheral blood sample (Fig. 5).

All genetic variants identified in the present study are described in Table I.

Table I

Summary of the genetic variants identified in the present study by NGS and Sanger sequencing analyses.

Table I

Summary of the genetic variants identified in the present study by NGS and Sanger sequencing analyses.

A, NGS analysis-OS tumor tissue
Gene IDChrPosRefAltTypeContextConsequencedbSNPCOSMICClinVarQual
JAK1chr165310489TCSNVCoding synonymous_variantrs2230588COSM3751351 100
ALKchr229416572TCSNVCoding missense_variantrs1670283 benign100
ALKchr229445458GTSNVCoding synonymous_variantrs3795850COSM5351767benign100
FGFR3chr41805799TCSNVIntronintron_variantrs3135888  100
FGFR3chr41807894GASNVCoding synonymous_variantrs7688609  100
PDGFRAchr455097835GCSNVIntronintron_variantrs4864504  100
FGFR4chr5176517326TCSNVIntronintron_variantrs422421  100
FGFR4chr5176517797CTSNVCoding missense_variantrs376618  100
FGFR4chr5176519516AGSNVIntron splice_region_variant, intron_variantrs3135925  100
FGFR4chr5176523562CASNVIntronintron_variantrs31777  100
FGFR4chr5176523597AGSNVIntron splice_region_variant, intron_variantrs31776  100
EGFRchr755219909GTSNVIntronintron_variantrs41364648  100
EGFRchr755228053ATSNVIntronintron_variantrs1558544  100
RETchr1043613843GTSNVCoding synonymous_variantrs1800861COSM4418405, COSM4418406benign100
KRASchr1225400206GTSNVIntronintron_variantrs10842518  100
B, Sanger sequencing (TP53 gene)-tumor tissue and peripheral blood
TP53 regionSampleTypeConsequence 
TP53 exon 5 deletionOS tumorDeletiondeletion 
TP53 c.712T>A (p.Cys238Ser)OS tumorSNVmissense variant 
TP53 c.215C>G (p.Pro72Arg)peripheral bloodSNVmissense variant 

[i] NGS, next-generation sequencing; OS, osteosarcoma; SNV, single nucleotide variant; dbSNP, Single Nucleotide Polymorphism Database.

Discussion

OS is the most common primary malignant bone tumor and it is characterized by its rare incidence and occurrence in the long bones of the extremities, notably in the femur. Adolescents and young adults are usually affected by the disease and improved 5-year overall survival rates are observed for younger patients (1). Molecular tests based on potential cancer-related genomic drivers could improve precision oncology for OS (21). However, knowledge about the genomic hallmarks related to OS etiology and progression is insufficient; thus, treatment options are still limited. In the present study, NGS analysis and Sanger sequencing were applied to investigate the genomic landscape of an adolescent patient diagnosed with OS and treated with a standard protocol. NGS analysis revealed that OS tumor sample harbored an intratumor heterogeneity signature and identified somatic variants in the following genes: JAK1, ALK, FGFR3, PDGFRA, FGFR4, EGFR, RET and KRAS.

Janus kinase (JAK) is a family of non-receptor tyrosine kinase proteins involved in the signal transduction of multiple cellular events, such as proliferation and differentiation (22,23). Some JAK mutations have been identified in different types of cancer. It has been reported that tumors with JAK1 mutations may exhibit high mutation burden and microsatellite instability, which may result in immune response alterations and contribute to tumor immune evasion (24). In the present study, a coding synonymous variant in the JAK1 gene (rs2230588; COSM3751351) was identified. Carvalho et al (25) reported the same JAK1 genetic variant in 25% (5/20) of tumor samples from patients diagnosed with head and neck squamous cell carcinoma.

Anaplastic lymphoma kinase (ALK) gene encodes a receptor tyrosine kinase and ALK mutations, rearrangements/fusions, or amplifications have been identified in several human cancers (26). ALK inhibitors represent an effective treatment strategy for patients with malignancies exhibiting ALK rearrangement and their effectiveness in patients with OS should be further explored. Ordulu et al (27) described a 73-year-old male with high-grade OS and lung metastases who showed sensitivity to ALK-targeted therapy harboring the EML4::ALK fusion and the ALKL1196M mutation. By contrast, Takeyasu et al (28) identified the ITSN2::ALK fusion in a 17-year-old male patient with OS showing poor sensitivity to alectinib therapy and progressive disease. In the present study, only DNA targets were investigated; therefore, gene fusions in RNA samples were not assessed. NGS analysis revealed a coding missense variant (rs1670283) and a synonymous variant (rs3795850; COSM5351767) in the ALK gene and both variants were classified as benign in the ClinVar database: https://www.ncbi.nlm.nih.gov/clinvar/variation/VCV000133472.35 and https://www.ncbi.nlm.nih.gov/clinvar/variation/VCV000335694.20, respectively. The ALK variant rs1670283 has also been associated with hereditary cancer-predisposing syndrome, breast cancer (29), anaplastic large cell lymphoma (30), gastric cancer (31) and benign tumor of the central nervous system (32). The ALK variant rs3795850 (COSM5351767) identified in the present study has also been associated with neuroblastoma susceptibility and detected in other cancer studies, including breast cancer (29), lung cancer (33,34) and Wilms tumor (35).

The fibroblast growth factor receptors (FGFRs) are a family of receptor tyrosine kinases involved in signaling pathways of different biological processes, in which the FGF/FGFR pathway plays a role in bone development and homeostasis (36,37). FGFR genomic alterations, including gene amplifications and gene mutations, have been described in OS (38,39). Additionally, clinical trials and case studies have evaluated the efficacy of FGFR inhibitors as a treatment option for patients with OS and reported improvements in progression-free survival (40,41). FGFR3 gene is a FGFR family member and FGFR3 mutations have been reported in several skeletal dysplasias (42-44). In the present study, the FGFR3 variants rs3135888 (intron variant) and rs7688609 (synonymous variant) were identified. By using NGS technology, Mansour et al also identified the FGFR3 rs7688609 variant in a patient with lung carcinoma (45). Kassem et al identified the FGFR3 rs7688609 variant in seven patients with brain tumors through liquid biopsy using NGS (46). The FGFR3 rs7688609 variant was also identified by panel-based NGS in a case of glioblastoma (47). FGFR4 also belongs to the FGFR family and the present study identified five FGFR4 variants: rs422421 (intron variant), rs376618 (missense variant), rs3135925 (splice region variant, intron variant), rs31777 (intron variant) and rs31776 (splice region variant, intron variant). In a meta-analysis study conducted by Moazeni-Roodi et al, the FGFR4 rs376618 variant was described in three studies and no association with overall cancer risk was found (48).

The platelet-derived growth factor receptor-alpha (PDGFRA) gene encodes a receptor tyrosine kinase involved in multiple cellular events. PDGFRA-mutated tumors can be found in several cancers and targeted inhibitors have been described as a treatment option to improve overall clinical outcomes (49-51). PDGFRA mutations commonly occur in the exons 12/14/18, notably in the exon 18 (D842V). Armstrong et al described a refractory metastatic OS case (7-year-old) with partial response to sorafenib (52). NGS technology revealed a PDGFRA D846V mutation in the initial tumor sample but not in the relapse sample, suggesting that this specific PDGFRA mutation is a sorafenib target. Using a custom NGS panel, a Brazilian study also identified molecular heterogeneity in OS tumor tissues, in which copy number variations (CNVs) were identified in the PDGFRA gene (53). In the present study, the intronic variant rs4864504 in the PDGFRA gene was identified.

The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase involved in downstream signaling cascades resulting in cellular growth and proliferation. EGFR deregulation has been identified in a number of cancers, especially lung cancer and frequently includes activating mutations in functional domains and amplifications (54,55). Treatment with EGFR tyrosine kinase inhibitors (TKIs) has emerged as an important strategy in cancer therapy to benefit patients harboring EGFR mutations (56). Two EGFR intronic variants (rs41364648 and rs1558544) were identified in the present study. Geißler et al (57) reported the EGFR rs1558544 variant in 7 of 25 patients with colorectal cancer. This EGFR rs1558544 variant was also found in a patient with cutaneous squamous cell carcinoma resistant to conventional treatments but successfully treated with anti-EGFR targeted therapy (58).

Rearranged during transfection (RET) is a proto-oncogene that encodes a transmembrane receptor tyrosine kinase involved in several cellular signaling pathways. RET gene mutations are commonly found in medullary thyroid carcinoma but can also be found in other cancers (59). Germline activating mutations and somatic amplifications in the RET gene have been described in patients with OS (60). The RET rs1800861 (COSM4418405) synonymous variant was identified in the present study. This RET variant was also found by NGS analysis in a patient with chronic myeloid leukemia (61).

Kirsten rat sarcoma viral oncogene homologue (KRAS) is a well-known proto-oncogene playing a central role as a signal transducer. KRAS mutations are commonly found in numerous human cancers and targeted therapies with KRAS inhibitors have emerged as promising treatment strategies (62). The uncommon KRAS intronic variant rs10842518 (g.25400206 G>T) was identified in the present study.

Most of the somatic SNVs described in the present study by NGS analysis were notably identified in genes encoding tyrosine kinase proteins, revealing an important intratumor heterogeneity signature that may contribute as an additional event for OS development. The application of TKIs in patients with OS has been evaluated as a strategy to achieve improved therapeutic efficacy, suggesting the simultaneous inhibition of several relevant receptor tyrosine kinases in OS (63,64).

TP53 is a tumor suppressor gene that encodes the p53 protein, which plays a key role in cell cycle control and genome integrity; thus, it is referred to as the guardian of the genome (65). Therefore, genetic alterations in the TP53 gene leading to its malfunction are hallmarks of several human cancers. Regarding OS, mutations in TP53 are commonly described (18,19,66). Additionally, OS is commonly diagnosed in Li-Fraumeni syndrome (LFS), a cancer predisposition syndrome characterized by inherited pathogenic germline variants in the TP53 gene (66,67). Mutational hotspots in the TP53 gene notably occur by nucleotide substitution in the coding sequence within exons 4-8(68). Wunder et al (69) investigated TP53 mutations (exons 4-10) in 196 OS tumors by single-strand conformation polymorphism and sequencing, identifying 38 mutations (19.4%) described as 23 missense mutations, 11 nonsense mutations, three splice site changes and one in-frame insertion. Chen et al (18) investigated somatic mutations in pediatric osteosarcoma samples by whole-genome sequencing and identified multiple somatic chromosomal alterations (notably structural variations) and SNVs. High rates of TP53 mutations, including TP53 rearrangements (50%; 16/32), missense mutations (22%; 7/32), nonsense mutations (16%; 5/32) and TP53 deletions (6%; 2/32), have been reported. By whole-genome sequencing, Ribi et al (70) identified patients with OS with a deletion in intron 1 of TP53, including exon 1, and a patient harboring a deletion, including the entire TP53 gene. By whole-exome sequencing, Bousquet et al (71) identified TP53 mutations in OS samples from three young patients, being described as STOP gained mutation, nonsynonymous mutation, deletions and splice site mutation. In the present study, no PCR products for TP53 exon 5 were detected in the tumor tissue sample, suggesting that a deletion event in exon 5 can lead to somatic TP53 inactivation and contribute to OS development. Sanger sequencing analysis revealed that the patient harbored the missense variant TP53 c.712T>A (p.Cys238Ser) in tumor tissue sample and the TP53 c.215C>G (p.Pro72Arg; rs1042522) germline missense variant in the peripheral blood sample. The variant TP53 c.712T>A (p.Cys238Ser) is reported to be pathogenic in the ClinVar database and was also identified in a family with LFS (72). The TP53 c.215C>G (p.Pro72Arg) is a common single nucleotide polymorphism and has been reported to be benign for LFS in the ClinVar database. Taken together, these findings reinforce the idea that TP53 mutations represent key oncogenic drivers in patients with OS.

The present study reported a case of an adolescent patient with OS harboring an intratumor heterogeneity signature. OS is a rare and challenging malignant tumor. In Brazil, patients diagnosed with OS are still treated with a standard protocol, reinforcing the need for genetic testing to guide diagnosis and treatment. The present study has several limitations. First, although the National Institute of Traumatology and Orthopedics is a reference center for musculoskeletal sarcomas care, DNA sequencing was not performed in all patients, which could improve the investigation of the association between the genetic findings and the development of pediatric OS or even variations in therapy response. Second, the present study faced the lack of diagnostic molecular genetic tests to help select patients at highest risk of disease progression for this NGS-based study. It was hypothesized that patients with OS at higher risk could harbor genetic alterations that contribute to both disease development and progression. Third, only DNA targets were investigated in the present study; therefore, the detection of clinically actionable gene fusions that have been described in OS was not assessed. Despite this, the findings of the present study had the potential to help to unravel part of the genetic landscape that constitutes the biologic heterogeneity of OS, providing extensive genomic information through a refined and advanced NGS-based technology. Additionally, all genetic variants identified in the present study had not previously been described in OS cases and might have contributed to disease development.

Notably, the number of genome-targeted therapies and the percentage of patients with cancer responding to these therapies have increased over the years (73). By contrast, patients with OS continue to be ineligible for targeted therapy due to lack of genetic testing in clinical routine, being a consequence of little investment and visibility, which notably represents a knowledge gap. As researchers, it was hypothesized that the present study facilitates understanding on the need for OS genetic investigation. In the next 5 years, it is hoped that more efforts with global collaboration initiatives in the scientific and medical communities will include genetic testing in clinical routine for OS and contribute to clinical trials for new treatment modalities with targeted therapies to achieve improved clinical responses. In conclusion, the present study added new information on genetic aspects contributing for OS development, especially in pediatric patients. Genomic testing for OS will represent the opportunity to identify potential cancer driver genes to aid clinical decision making. Therefore, genetic profiling of OS needs further investigation.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The NGS datasets generated and/or analyzed during the current study are available in Figshare (https://doi.org/10.6084/m9.figshare.26015227.v1). Sanger sequencing datasets for TP53 (exons 4-8) in the current study are available in Figshare (https://doi.org/10.6084/m9.figshare.27115351.v1).

Authors' contributions

MJ contributed to the conception of the present study, performed the experiments, analyzed the molecular data and wrote the original and final draft of the manuscript. RS and TC contributed significantly to the NGS experiments and analysis. EL and RP contributed to the acquisition and writing of the clinical data. AC contributed to the acquisition and writing of the histopathological data. AL and WM analyzed all the clinical data. GA and MO reviewed the study and confirmed the authenticity of all the raw data. All the authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics and Research Committee of the National Institute of Traumatology and Orthopedics (approval no. CAAE: 60632822.4.0000.5273). Written informed consent was obtained from the patient and from the patient's legal guardian.

Patient consent for publication

Written informed consent was obtained from the patient and from her legal guardian.

Competing interests

The authors declare that they have no competing interests.

Authors' information

MJ: https://orcid.org/0000-0002-7351-5588

RS: https://orcid.org/0000-0003-2009-537X

TC: https://orcid.org/0009-0005-9493-1352

EL: https://orcid.org/0000-0002-1111-4906

RP: https://orcid.org/0000-0001-7837-5134

GA: https://orcid.org/0000-0003-2246-719X

MO: https://orcid.org/0000-0002-2983-9593

References

1 

Lee JA, Lim J, Jin HY, Park M, Park HJ, Park JW, Kim JH, Kang HG and Won YJ: Osteosarcoma in adolescents and young adults. Cells. 10(2684)2021.PubMed/NCBI View Article : Google Scholar

2 

Ottaviani G and Jaffe N: The epidemiology of osteosarcoma. Cancer Treat Res. 152:3–13. 2009.PubMed/NCBI View Article : Google Scholar

3 

Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023.PubMed/NCBI View Article : Google Scholar

4 

Balach T, Stacy GS and Peabody TD: The clinical evaluation of bone tumors. Radiol Clin North Am. 49:1079–1093. 2011.PubMed/NCBI View Article : Google Scholar

5 

Carrle D and Bielack SS: Current strategies of chemotherapy in osteosarcoma. Int Orthop. 30:445–451. 2006.PubMed/NCBI View Article : Google Scholar

6 

Zoumpoulidou G, Alvarez-Mendoza C, Mancusi C, Ahmed RM, Denman M, Steele CD, Tarabichi M, Roy E, Davies LR, Manji J, et al: Therapeutic vulnerability to PARP1,2 inhibition in RB1-mutant osteosarcoma. Nat Commun. 12(7064)2021.PubMed/NCBI View Article : Google Scholar

7 

Astolfi A, Nannini M, Indio V, Schipani A, Rizzo A, Perrone AM, De Iaco P, Pirini MG, De Leo A, Urbini M, et al: Genomic database analysis of uterine leiomyosarcoma mutational profile. Cancers (Basel). 12(2126)2020.PubMed/NCBI View Article : Google Scholar

8 

Rizzo A, Pantaleo MA, Saponara M and Nannini M: Current status of the adjuvant therapy in uterine sarcoma: A literature review. World J Clin Cases. 7:1753–1763. 2019.PubMed/NCBI View Article : Google Scholar

9 

Rizzo A, Nannini M, Astolfi A, Indio V, De Iaco P, Perrone AM, De Leo A, Incorvaia L, Di Scioscio V and Pantaleo MA: Impact of chemotherapy in the adjuvant setting of early stage uterine leiomyosarcoma: A systematic review and updated meta-analysis. Cancers (Basel). 12(1899)2020.PubMed/NCBI View Article : Google Scholar

10 

Boye K, Longhi A, Guren T, Lorenz S, Næss S, Pierini M, Taksdal I, Lobmaier I, Cesari M, Paioli A, et al: Pembrolizumab in advanced osteosarcoma: Results of a single-arm, open-label, phase 2 trial. Cancer Immunol Immunother. 70:2617–2624. 2021.PubMed/NCBI View Article : Google Scholar

11 

Davis KL, Fox E, Merchant MS, Reid JM, Kudgus RA, Liu X, Minard CG, Voss S, Berg SL, Weigel BJ and Mackall CL: Nivolumab in children and young adults with relapsed or refractory solid tumours or lymphoma (ADVL1412): A multicentre, open-label, single-arm, phase 1-2 trial. Lancet Oncol. 21:541–550. 2020.PubMed/NCBI View Article : Google Scholar

12 

Monga V, Dodd R, Scherer A, Gutierrez WR, Tanas M, Mott SL and Milhem M: Phase Ib study of decitabine in combination with gemcitabine in treatment of advanced soft tissue and bone sarcomas. J Clin Oncol. 38:11550. 2020.

13 

Krishnadas DK, Shusterman S, Bai F, Diller L, Sullivan JE, Cheerva AC, George RE and Lucas KG: A phase I trial combining decitabine/dendritic cell vaccine targeting MAGE-A1, MAGE-A3 and NY-ESO-1 for children with relapsed or therapy-refractory neuroblastoma and sarcoma. Cancer Immunol Immunother. 64:1251–1260. 2015.PubMed/NCBI View Article : Google Scholar

14 

Chi SN, Yi JS, Williams PM, Roy-Chowdhuri S, Patton DR, Coffey BD, Reid JM, Piao J, Saguilig L, Alonzo TA, et al: Tazemetostat in patients with tumors with alterations in EZH2 or the SWI/SNF complex: Results from NCI-COG pediatric MATCH trial Arm C (APEC1621C). J Clin Oncol. 40 (16 Suppl)(10009)2022.

15 

Hanahan D: Hallmarks of cancer: New dimensions. Cancer Discov. 12:31–46. 2022.PubMed/NCBI View Article : Google Scholar

16 

Schwartzberg L, Kim ES, Liu D and Schrag D: Precision oncology: Who, how, what, when, and when not? Am Soc Clin Oncol Educ Book. 37:160–169. 2017.PubMed/NCBI View Article : Google Scholar

17 

Gagan J and Van Allen EM: Next-generation sequencing to guide cancer therapy. Genome Med. 7(80)2015.PubMed/NCBI View Article : Google Scholar

18 

Chen X, Bahrami A, Pappo A, Easton J, Dalton J, Hedlund E, Ellison D, Shurtleff S, Wu G, Wei L, et al: Recurrent somatic structural variations contribute to tumorigenesis in pediatric osteosarcoma. Cell Rep. 7:104–112. 2014.PubMed/NCBI View Article : Google Scholar

19 

Czarnecka AM, Synoradzki K, Firlej W, Bartnik E, Sobczuk P, Fiedorowicz M, Grieb P and Rutkowski P: Molecular biology of osteosarcoma. Cancers (Basel). 12(2130)2020.PubMed/NCBI View Article : Google Scholar

20 

Leite C, Delmonico L, Alves G, Gomes RJ, Martino MR, Silva AR, Moreira ADS, Maioli MC, Scherrer LR, Bastos EF, et al: Screening of mutations in the additional sex combs like 1, transcriptional regulator, tumor protein p53, and KRAS proto-oncogene, GTPase/NRAS proto-oncogene, GTPase genes of patients with myelodysplastic syndrome. Biomed Rep. 7:343–348. 2017.PubMed/NCBI View Article : Google Scholar

21 

Pestana RC, Groisberg R, Roszik J and Subbiah V: Precision oncology in sarcomas: Divide and conquer. JCO Precis Oncol. 3(PO.18.00247)2019.PubMed/NCBI View Article : Google Scholar

22 

Bousoik E and Aliabadi HM: ‘Do We Know Jack’ about JAK? A closer look at JAK/STAT signaling pathway. Front Oncol. 8(287)2018.PubMed/NCBI View Article : Google Scholar

23 

Hu X, Li J, Fu M, Zhao X and Wang W: The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct Target Ther. 6(402)2021.PubMed/NCBI View Article : Google Scholar

24 

Albacker LA, Wu J, Smith P, Warmuth M, Stephens PJ, Zhu P, Yu L and Chmielecki J: Loss of function JAK1 mutations occur at high frequency in cancers with microsatellite instability and are suggestive of immune evasion. PLoS One. 12(e0176181)2017.PubMed/NCBI View Article : Google Scholar

25 

Carvalho TG, Carvalho AC, Maia DCC, Ogawa JK, Carvalho AL and Vettore AL: Search for mutations in signaling pathways in head and neck squamous cell carcinoma. Oncol Rep. 30:334–340. 2013.PubMed/NCBI View Article : Google Scholar

26 

Corte CMD, Viscardi G, Liello R, Fasano M, Martinelli E, Troiani T, Ciardiello F and Morgillo F: Role and targeting of anaplastic lymphoma kinase in cancer. Mol Cancer. 17(30)2018.PubMed/NCBI View Article : Google Scholar

27 

Ordulu Z, Giunta P, Hung WT, Hung YP, Simon J, Fintelmann FJ, Lennerz JK, Naxerova K and Cote GM: Sensitivity to ALK-directed therapy in osteosarcoma with an acquired ALK rearrangement. JCO Precis Oncol. 7(e2300287)2023.PubMed/NCBI View Article : Google Scholar

28 

Takeyasu Y, Okuma HS, Kojima Y, Nishikawa T, Tanioka M, Sudo K, Shimoi T, Noguchi E, Arakawa A, Mori T, et al: Impact of ALK inhibitors in patients with ALK-rearranged nonlung solid tumors. JCO Precis Oncol. 5(PO.20.00383)2021.PubMed/NCBI View Article : Google Scholar

29 

Pan ZW, Wang XJ, Chen T, Ding XW, Jiang X, Gao Y, Mo WJ, Huang Y, Lou CJ and Cao WM: Deleterious mutations in DNA repair gene FANCC exist in BRCA1/2-negative Chinese familial breast and/or ovarian cancer patients. Front Oncol. 9(169)2019.PubMed/NCBI View Article : Google Scholar

30 

Noguchi K, Ikawa Y, Takenaka M, Sakai Y, Fujiki T, Kuroda R, Ikeda H, Abe T, Sakai S and Wada T: Acquired L1196M ALK mutation in anaplastic lymphoma kinase-positive anaplastic large cell lymphoma during alectinib administration. EJHaem. 4:305–308. 2023.PubMed/NCBI View Article : Google Scholar

31 

Machlowska J, Kapusta P, Baj J, Morsink FHM, Wołkow P, Maciejewski R, Offerhaus GJA and Sitarz R: High-throughput sequencing of gastric cancer patients: Unravelling genetic predispositions towards an early-onset subtype. Cancers (Basel). 12(1981)2020.PubMed/NCBI View Article : Google Scholar

32 

Taher MM, Hassan AA, Saeed M, Jastania RA, Nageeti TH, Alkhalidi H, Dairi G, Abduljaleel Z, Athar M, Bouazzaoui A, et al: Next generation DNA sequencing of atypical choroid plexus papilloma of brain: Identification of novel mutations in a female patient by Ion Proton. Oncol Lett. 18:5063–5076. 2019.PubMed/NCBI View Article : Google Scholar

33 

Choi YL, Soda M, Yamashita Y, Ueno T, Takashima J, Nakajima T, Yatabe Y, Takeuchi K, Hamada T, Haruta H, et al: EML4-ALK mutations in lung cancer that confer resistance to ALK inhibitors. N Engl J Med. 363:1734–1739. 2010.PubMed/NCBI View Article : Google Scholar

34 

Doebele RC, Pilling AB, Aisner DL, Kutateladze TG, Le AT, Weickhardt AJ, Kondo KL, Linderman DJ, Heasley LE, Franklin WA, et al: Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin Cancer Res. 18:1472–1482. 2012.PubMed/NCBI View Article : Google Scholar

35 

Wegert J, Ishaque N, Vardapour R, Geörg C, Gu Z, Bieg M, Ziegler B, Bausenwein S, Nourkami N, Ludwig N, et al: Mutations in the SIX1/2 pathway and the DROSHA/DGCR8 miRNA microprocessor complex underlie high-risk blastemal type Wilms tumors. Cancer Cell. 27:298–311. 2015.PubMed/NCBI View Article : Google Scholar

36 

Dai S, Zhou Z, Chen Z, Xu G and Chen Y: Fibroblast growth factor receptors (FGFRs): Structures and small molecule inhibitors. Cells. 8(614)2019.PubMed/NCBI View Article : Google Scholar

37 

Su N, Jin M and Chen L: Role of FGF/FGFR signaling in skeletal development and homeostasis: Learning from mouse models. Bone Res. 2(14003)2014.PubMed/NCBI View Article : Google Scholar

38 

Barøy T, Chilamakuri CSR, Lorenz S, Sun J, Bruland ØS, Myklebost O and Meza-Zepeda LA: Genome analysis of osteosarcoma progression samples identifies FGFR1 overexpression as a potential treatment target and CHM as a candidate tumor suppressor gene. PLoS One. 11(e0163859)2016.PubMed/NCBI View Article : Google Scholar

39 

Brown LM, Ekert PG and Fleuren EDG: Biological and clinical implications of FGFR aberrations in paediatric and young adult cancers. Oncogene. 42:1875–1888. 2023.PubMed/NCBI View Article : Google Scholar

40 

Davis LE, Bolejack V, Ryan CW, Ganjoo KN, Loggers ET, Chawla S, Agulnik M, Livingston MB, Reed D, Keedy V, et al: Randomized double-blind phase II study of Regorafenib in patients with metastatic osteosarcoma. J Clin Oncol. 37:1424–1431. 2019.PubMed/NCBI View Article : Google Scholar

41 

Duffaud F, Mir O, Boudou-Rouquette P, Piperno-Neumann S, Penel N, Bompas E, Delcambre C, Kalbacher E, Italiano A, Collard O, et al: Efficacy and safety of regorafenib in adult patients with metastatic osteosarcoma: A non-comparative, randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol. 20:120–133. 2019.PubMed/NCBI View Article : Google Scholar

42 

Du X, Xie Y, Xian CJ and Chen L: Role of FGFs/FGFRs in skeletal development and bone regeneration. J Cell Physiol. 227:3731–3743. 2012.PubMed/NCBI View Article : Google Scholar

43 

Xue Y, Sun A, Mekikian PB, Martin J, Rimoin DL, Lachman RS and Wilcox WR: FGFR3 mutation frequency in 324 cases from the international skeletal dysplasia registry. Mol Genet Genomic Med. 2:497–503. 2014.PubMed/NCBI View Article : Google Scholar

44 

Wen X, Li X, Tang Y, Tang J, Zhou S, Xie Y, Guo J, Yang J, Du X, Su N and Chen L: Chondrocyte FGFR3 regulates bone mass by inhibiting osteogenesis. J Biol Chem. 291:24912–24921. 2016.PubMed/NCBI View Article : Google Scholar

45 

Mansour H, Ouhajjou A, Bajic VB and Incitti R: Next-Generation sequencing at high sequencing depth as a tool to study the evolution of metastasis driven by genetic change events of lung squamous cell carcinoma. Front Oncol. 10(1215)2020.PubMed/NCBI View Article : Google Scholar

46 

Kassem NM, Kassem HA, Selim H and Hafez M: Targeted next generation sequencing provides insight for the genetic alterations in liquid biopsy of Egyptian brain tumor patients. Egypt J Med Hum Genet. 23(23)2022.

47 

Loriguet L, Morisse MC, Dremaux J, Collet L, Attencourt C, Coutte A, Boone M, Sevestre H, Galmiche A, Gubler B, et al: Combining genomic analyses with tumour-derived slice cultures for the characterization of an EGFR-activating kinase mutation in a case of glioblastoma. BMC Cancer. 18(964)2018.PubMed/NCBI View Article : Google Scholar

48 

Moazeni-Roodi A, Sarabandi S, Karami S, Hashemi M and Ghavami S: An updated meta-analysis of the association between fibroblast growth factor receptor 4 polymorphisms and susceptibility to cancer. Biosci Rep. 40(BSR20192051)2020.PubMed/NCBI View Article : Google Scholar

49 

Sun Y, Yue L, Xu P and Hu W: An overview of agents and treatments for PDGFRA-mutated gastrointestinal stromal tumors. Front Oncol. 12(927587)2022.PubMed/NCBI View Article : Google Scholar

50 

Velghe AI, Cauwenberghe SV, Polyansky AA, Chand D, Montano-Almendras CP, Charni S, Hallberg B, Essaghir A and Demoulin JB: PDGFRA alterations in cancer: Characterization of a gain-of-function V536E transmembrane mutant as well as loss-of-function and passenger mutations. Oncogene. 33:2568–2576. 2014.PubMed/NCBI View Article : Google Scholar

51 

Wagner AJ, Kindler H, Gelderblom H, Schöffski P, Bauer S, Hohenberger P, Kopp HG, Lopez-Martin JA, Peeters M, Reichardt P, et al: A phase II study of a human anti-PDGFRα monoclonal antibody (olaratumab, IMC-3G3) in previously treated patients with metastatic gastrointestinal stromal tumors. Ann Oncol. 28:541–546. 2017.PubMed/NCBI View Article : Google Scholar

52 

Armstrong AE, Walterhouse DO, Leavey PJ, Reichek J and Walz AL: Prolonged response to sorafenib in a patient with refractory metastatic osteosarcoma and a somatic PDGFRA D846V mutation. Pediatr Blood Cancer. 66(e27493)2019.PubMed/NCBI View Article : Google Scholar

53 

Guimarães GM, Tesser-Gamba F, Petrilli AS, Donato-Macedo CRP, Alves MTS, de Lima FT, Garcia-Filho RJ, Oliveira R and Toledo SRC: Molecular profiling of osteosarcoma in children and adolescents from different age groups using a next-generation sequencing panel. Cancer Genet. 258-259:85–92. 2021.PubMed/NCBI View Article : Google Scholar

54 

Jin R, Peng L, Shou J, Wang J, Jin Y, Liang F, Zhao J, Wu M, Li Q, Zhang B, et al: EGFR-Mutated squamous cell lung cancer and its association with outcomes. Front Oncol. 11(680804)2021.PubMed/NCBI View Article : Google Scholar

55 

Liu H, Zhang B and Sun Z: Spectrum of EGFR aberrations and potential clinical implications: Insights from integrative pan-cancer analysis. Cancer Commun (Lond). 40:43–59. 2020.PubMed/NCBI View Article : Google Scholar

56 

Nan X, Xie C, Yu X and Liu J: EGFR TKI as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer. Oncotarget. 8:75712–75726. 2017.PubMed/NCBI View Article : Google Scholar

57 

Geißler AL, Geißler M, Kottmann D, Lutz L, Fichter CD, Fritsch R, Weddeling B, Makowiec F, Werner M and Lassmann S: ATM mutations and E-cadherin expression define sensitivity to EGFR-targeted therapy in colorectal cancer. Oncotarget. 8:17164–17190. 2017.PubMed/NCBI View Article : Google Scholar

58 

Capalbo C, Belardinilli F, Filetti M, Parisi C, Petroni M, Colicchia V, Tessitore A, Santoni M, Coppa A, Giannini G and Marchetti P: Effective treatment of a platinum-resistant cutaneous squamous cell carcinoma case by EGFR pathway inhibition. Mol Clin Oncol. 9:30–34. 2018.PubMed/NCBI View Article : Google Scholar

59 

Verrienti A, Grani G, Sponziello M, Pecce V, Damante G, Durante C, Russo D and Filetti S: Precision oncology for RET-related tumors. Front Oncol. 12(992636)2022.PubMed/NCBI View Article : Google Scholar

60 

Kovac M, Woolley C, Ribi S, Blattmann C, Roth E, Morini M, Kovacova M, Ameline B, Kulozik A, Bielack S, et al: Germline RET variants underlie a subset of paediatric osteosarcoma. J Med Genet. 58:20–24. 2021.PubMed/NCBI View Article : Google Scholar

61 

Sklarz LM, Wittke C, Krohn S, GROßE-Thie C, Junghanss C, Escobar HM and Glaeser H: Genetic mutations in a patient with chronic myeloid leukemia showing blast crisis 10 years after presentation. Anticancer Res. 38:3961–3966. 2018.PubMed/NCBI View Article : Google Scholar

62 

Huang L, Guo Z, Wang F and Fu L: KRAS mutation: From undruggable to druggable in cancer. Signal Transduct Target Ther. 6(386)2021.PubMed/NCBI View Article : Google Scholar

63 

Chen C, Shi Q, Xu J, Ren T, Huang Y and Guo W: Current progress and open challenges for applying tyrosine kinase inhibitors in osteosarcoma. Cell Death Discov. 8(488)2022.PubMed/NCBI View Article : Google Scholar

64 

Tian Z, Niu X and Yao W: Receptor tyrosine kinases in osteosarcoma treatment: Which is the key target? Front Oncol. 10(1642)2020.PubMed/NCBI View Article : Google Scholar

65 

Borrero LJH and El-Deiry WS: Tumor suppressor p53: Biology, signaling pathways, and therapeutic targeting. Biochim Biophys Acta Rev Cancer. 1876(188556)2021.PubMed/NCBI View Article : Google Scholar

66 

Mirabello L, Yeager M, Mai PL, Gastier-Foster JM, Gorlick R, Khanna C, Patiño-Garcia A, Sierrasesúmaga L, Lecanda F, Andrulis IL, et al: Germline TP53 variants and susceptibility to osteosarcoma. J Natl Cancer Inst. 107(djv101)2015.PubMed/NCBI View Article : Google Scholar

67 

Hameed M and Mandelker D: Tumor syndromes predisposing to osteosarcoma. Adv Anat Pathol. 25:217–222. 2018.PubMed/NCBI View Article : Google Scholar

68 

Petitjean A, Mathe E, Kato S, Ishioka C, Tavtigian SV, Hainaut P and Olivier M: Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat. 28:622–629. 2007.PubMed/NCBI View Article : Google Scholar

69 

Wunder JS, Gokgoz N, Parkes R, Bull SB, Eskandarian S, Davis AM, Beauchamp CP, Conrad EU, Grimer RJ, Healey JH, et al: TP53 mutations and outcome in osteosarcoma: a prospective, multicenter study. J Clin Oncol. 23:1483–1490. 2005.PubMed/NCBI View Article : Google Scholar

70 

Ribi S, Baumhoer D, Lee K, Edison Teo AS, Madan B, Zhang K, Kohlmann WK, Yao F, Lee WH, et al: TP53 intron 1 hotspot rearrangements are specific to sporadic osteosarcoma and can cause Li-Fraumeni syndrome. Oncotarget. 6:7727–7740. 2015.PubMed/NCBI View Article : Google Scholar

71 

Bousquet M, Noirot C, Accadbled F, de Gauzy JS, Castex MP, Brousset P and Gomez-Brouchet A: Whole-exome sequencing in osteosarcoma reveals important heterogeneity of genetic alterations. Ann Oncol. 27:738–744. 2016.PubMed/NCBI View Article : Google Scholar

72 

Balmaña J, Nomdedéu J, Díez O, Sabaté JM, Balil A, Pericay C, López JJ, Brunet J, Baiget M and Alonso C: Description of a new TP53 gene germline mutation in a family with the Li-Fraumeni syndrome. Genetic counselling to healthy mutation carriers. Med Clin (Barc). 119:497–499. 2002.PubMed/NCBI View Article : Google Scholar : (In Spanish).

73 

Haslam A, Kim MS and Prasad V: Updated estimates of eligibility for and response to genome-targeted oncology drugs among US cancer patients, 2006-2020. Ann Oncol. 32:926–932. 2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-2025
Volume 22 Issue 3

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chantre‑Justino M, Silvestre RT, De Castro TL, Luz E, Pinheiro RD, Caruso A, Lopes AD, Meohas W, Alves G, Ornellas MF, Ornellas MF, et al: Genetic profiling of osteosarcoma in an adolescent using a next‑generation sequencing panel and Sanger sequencing: A case report and review of the literature. Biomed Rep 22: 42, 2025.
APA
Chantre‑Justino, M., Silvestre, R.T., De Castro, T.L., Luz, E., Pinheiro, R.D., Caruso, A. ... Ornellas, M.F. (2025). Genetic profiling of osteosarcoma in an adolescent using a next‑generation sequencing panel and Sanger sequencing: A case report and review of the literature. Biomedical Reports, 22, 42. https://doi.org/10.3892/br.2025.1920
MLA
Chantre‑Justino, M., Silvestre, R. T., De Castro, T. L., Luz, E., Pinheiro, R. D., Caruso, A., Lopes, A. D., Meohas, W., Alves, G., Ornellas, M. F."Genetic profiling of osteosarcoma in an adolescent using a next‑generation sequencing panel and Sanger sequencing: A case report and review of the literature". Biomedical Reports 22.3 (2025): 42.
Chicago
Chantre‑Justino, M., Silvestre, R. T., De Castro, T. L., Luz, E., Pinheiro, R. D., Caruso, A., Lopes, A. D., Meohas, W., Alves, G., Ornellas, M. F."Genetic profiling of osteosarcoma in an adolescent using a next‑generation sequencing panel and Sanger sequencing: A case report and review of the literature". Biomedical Reports 22, no. 3 (2025): 42. https://doi.org/10.3892/br.2025.1920