International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Osteosarcoma is a highly aggressive primary bone malignancy characterized by the production of osteoid and bone tissue, predominantly affecting the metaphyseal regions of long bones, particularly the distal femur, proximal tibia and proximal humerus (1). It generally arises in adolescents and young adults, peaking between the ages of 10 and 20 years, during periods of rapid skeletal growth (1). Epidemiologically, osteosarcoma has an annual incidence of ~3-4 cases per million individuals in the general population, with a notable male predominance (~1.5:1) (2). Geographic variations in incidence have been observed, with certain studies indicating higher rates in African populations and lower frequencies in Asian cohorts (2). Recent advancements in genomic studies have identified possible genetic risk factors, including mutations in the tumor protein 53 (TP53) gene linked to Li-Fraumeni syndrome (3,4). Furthermore, previous exposure to radiation and certain chemotherapeutic agents have been implicated in the etiology of osteosarcoma. Understanding these epidemiological patterns and genetic predispositions is crucial for developing targeted screening and prevention strategies in at-risk populations (5).
The current standard of care for osteosarcoma primarily involves a multidisciplinary approach, combining neoadjuvant chemotherapy, surgical resection and adjuvant chemotherapy (6). High-dose multi-agent chemotherapy, typically including doxorubicin, cisplatin and methotrexate, aims to shrink tumors before surgery and eliminate micrometastatic disease postoperatively (7). However, despite advances in treatment, outcomes remain suboptimal, with a 5-year survival rate of ~60-70% for localized disease and significantly lower for metastatic cases. Limitations in treatment efficacy arise from tumor heterogeneity, development of chemoresistance and late diagnosis (8). Furthermore, the potential for severe adverse effects from aggressive chemotherapy regimens poses significant challenges to patient quality of life (9). Emerging strategies that incorporate multi-omics analyses are being explored to identify novel therapeutic targets and improve individualized treatment plans.
Multi-omics is an integrative approach that combines various layers of biological information, such as genomics, transcriptomics, proteomics and metabolomics, to gain comprehensive insights into cancer biology (10). This concept facilitates the understanding of the complex molecular interplay driving tumorigenesis, progression and response to therapy (11). By simultaneously analyzing the genetic profiles (including gene mutations and epigenetic changes), gene (mRNA) expression, protein expression, other RNA expression and metabolic profiles of tumors, researchers can identify novel biomarkers and therapeutic targets that may be overlooked by traditional methods such as isolated genomic, transcriptomic or proteomic analyses (12). Recent studies have highlighted the potential of multi-omics to refine patient stratification and personalize treatment strategies, leading to improved outcomes in cancers like osteosarcoma (4,13). The advent of advanced technologies and data analytics further enhances the capacity to integrate these diverse data types, positioning multi-omics as a cornerstone of precision medicine in oncology (14).
The purpose of this review is to synthesize current advances in multi-omics technologies and their application in osteosarcoma research, highlighting how these integrative approaches can enhance the understanding of tumor biology, improve prognostic stratification and inform the development of personalized treatment strategies for achieving better patient outcomes.
The genetic landscape of osteosarcoma is characterized by multiple recurrent mutations and dysregulated pathways that contribute to its pathogenesis (15). Key mutations commonly observed in osteosarcoma include alterations in the TP53, retinoblastoma protein 1 (RB1) and mouse double minute 2 (MDM2) genes (Fig. 1) (16-18). TP53, a well-established tumor suppressor gene, is frequently mutated in numerous cancers, including osteosarcoma, leading to the loss of its normal function in cell cycle regulation and apoptosis (19). Mutations in RB1, another critical tumor suppressor, disrupt the Rb-E2F pathway, promoting uncontrolled cellular proliferation and contributing to tumor growth (20). Meanwhile, MDM2 overexpression results in the degradation of p53, further exacerbating the dysregulation of apoptosis and cell survival (21).
These genetic alterations not only inform tumor behavior but also influence treatment response. For example, osteosarcoma tumors with TP53 mutations may exhibit resistance to conventional chemotherapy agents, necessitating alternative therapeutic approaches (22). Furthermore, specific molecular subtypes identified through genomic profiling can be targeted with novel therapeutic agents, emphasizing the importance of personalizing treatment based on the genetic characteristics of the tumor (23).
The TME plays a crucial role in shaping the behavior and progression of osteosarcoma (24). Comprising various cell types, including immune cells, fibroblasts, endothelial cells and extracellular matrix (ECM) components, the TME significantly influences tumor growth and metastasis (Fig. 1) (25). Interactions between osteosarcoma cells and immune cells, such as macrophages and T-cells, can modulate the immune response, either promoting tumor progression through immune evasion or enhancing anti-tumor activity (26).
Certain soluble factors within the TME can also affect the behavior of osteosarcoma cells (27). For instance, the release of cytokines and growth factors from stromal cells can promote angiogenesis and tumor-supportive inflammation, creating a favorable niche for tumor growth (28). Additionally, components of the ECM, such as collagen and fibronectin, can interact with integrins on the surface of osteosarcoma cells, triggering signaling pathways that enhance proliferation and migration (29). Understanding the complex interplay between osteosarcoma cells and their microenvironment holds potential for therapeutic intervention. Strategies aimed at modulating the TME, such as targeted therapies that disrupt tumor-stroma interactions or immune checkpoint inhibitors, are being explored in ongoing clinical trials (30).
Tumor heterogeneity is a hallmark feature of osteosarcoma, characterized by variations in the cellular composition, genetic makeup and functional properties of the tumor (31). Histopathological analyses often reveal a mixture of osteoblastic, chondroblastic and fibroblastic cell types within osteosarcoma, reflecting the diverse lineage origins and differentiation states of the tumor (31).
This heterogeneity has profound implications for treatment resistance, as subpopulations of cells may exhibit differential responses to therapy. For instance, a subset of cancer stem-like cells has been identified in osteosarcoma that is thought to be responsible for tumor recurrence and metastasis due to their enhanced survival capabilities and resistance to conventional therapies (32). The presence of these resistant subpopulations can lead to incomplete responses to treatment, ultimately resulting in unfavorable prognostic outcomes (33).
Furthermore, the dynamic nature of tumor evolution complicates the understanding of treatment resistance (34). As tumors are exposed to therapeutic pressures, they may undergo clonal selection, leading to the emergence of resistant variants that contribute to disease treatment resistance progression (35). Integrating multi-omics approaches to profile the genetic, transcriptomic and proteomic landscapes of osteosarcoma will provide insights into the mechanisms of heterogeneity and resistance, paving the way for the development of targeted therapeutic strategies tailored to specific tumor characteristics (36).
The complexity of osteosarcoma is a multifaceted challenge that necessitates a deeper understanding of its genetic landscape, the role of the TME and the heterogeneity within the tumor itself (37). Advances in multi-omics technologies hold promise for elucidating the underlying mechanisms driving osteosarcoma biology and can aid in the development of innovative therapeutic strategies that enhance patient outcomes (38). As we move towards precision medicine, a thorough comprehension of these complexities will be indispensable in addressing the limitations of current treatment paradigms.
Recent advances in whole-genome sequencing (WGS) have significantly enhanced our understanding of the genomic landscape of osteosarcoma (39). WGS allows for the identification of genetic alterations, comprehensively analyzing both coding and non-coding regions of the genome (40). Studies have identified critical driver mutations in key genes such as TP53 (16), RB1 (41) and MDM2 (42), which play pivotal roles in tumor development and progression (Table I).
Mutations in the TP53 gene are frequently observed in osteosarcoma, which are critical for tumor progression (22). These mutations lead to the inactivation of its tumor suppressor function, compromising the cellular mechanisms of apoptosis and DNA repair (22). A comprehensive study has established that germline TP53 mutations significantly increase susceptibility to osteosarcoma and correlate with poorer survival outcomes (16). The RB1 is another crucial tumor suppressor associated with osteosarcoma. Deletions or mutations in RB1, frequently observed in conjunction with TP53 mutations, enhance cell cycle progression and contribute to oncogenesis (41). It is noteworthy that the co-mutation of TP53 and RB1 is often linked to aggressive disease phenotypes and metastasis (17).
Amplification of the MDM2 gene, which encodes a protein that inhibits TP53, is present in ~10-15% of osteosarcoma cases. Increased MDM2 expression facilitates the bypass of TP53-mediated apoptosis, aiding in tumor growth and resistance to therapies (42-44). Its role in the differentiation of osteosarcoma from benign lesions highlights its diagnostic significance (45). The PI3K/AKT signaling pathway is frequently activated in osteosarcoma. For instance, a study on C-type lectin domain family 3 member A demonstrated that its suppression led to reduced cell proliferation through the AKT serine/threonine kinase 1 (AKT1)/mammalian target of rapamycin (mTOR) axis (46). Furthermore, ménage à trios 1 has been shown to facilitate lung metastasis via upregulation of AKT1 expression, underscoring the pathway's role in metastatic spread (47).
The fibroblast growth factor receptor (FGFR) signaling pathway has gained attention due to its involvement in osteosarcoma progression (48). The FGFR signaling pathway, particularly FGFR1, plays a critical role in the progression of osteosarcoma. Overexpression of FGF2 and FGFRs has been linked to increased cell migration, invasion and metastasis, notably through intercellular adhesion molecule-1 expression (49). Furthermore, nuclear FGFR1 contributes to radiation resistance by inducing cell survival mechanisms, such as G-2 checkpoint adaptation and histone modifications. Targeting FGFR signaling may not only hinder tumor progression but also enhance the efficacy of radiation therapy in osteosarcoma, providing a promising therapeutic strategy (50).
Aberrant epigenetic modifications have been identified in osteosarcoma that may contribute to its aggressiveness (51). For instance, alterations in histone markers, such as H3K27me3, have been noted in various osteosarcoma subtypes, indicating heterogeneous disease behavior (52). Such findings emphasize the need for integrating epigenetic profiling into the genomic landscape of osteosarcoma. Recent genomic profiling has identified fusion genes, such as fibronectin 1-FGFR1, which present new therapeutic targets (53). Such fusions indicate a need for targeted sequencing in osteosarcoma patients, particularly those with refractory disease, as they may provide insights into novel treatment strategies.
The integration of genomic findings into clinical practice presents an opportunity for personalized medicine in osteosarcoma. Genetic profiling can direct the selection of targeted therapies, optimize chemotherapeutic regimens and enhance the monitoring of disease progression. For instance, the detection of MDM2 overexpression may prompt the use of specific MDM2 inhibitors, tailoring treatment to the genetic landscape of the tumor (43,45).
The totality of genomic research on osteosarcoma illustrates a landscape of intricate genetic alterations that drive tumorigenesis (54). The mutations in tumor suppressor genes like TP53 and RB1, the amplification of oncogenes such as MDM2 and AKT1, and disturbances to growth factor signaling pathways underscore the heterogeneous nature of this malignancy (16,41,42,47). Advancements in understanding these changes are crucial not only in elucidating osteosarcoma biology but also in providing a framework for developing targeted therapies and precision medicine approaches. Future research should focus on synthesizing these genomic insights with clinical outcomes to enhance treatment efficacy and patient survival rates.
The application of transcriptomics in the study of osteosarcoma has unveiled significant insights into the molecular mechanisms underlying this aggressive bone tumor (55). By analyzing various RNA types, particularly messenger RNA, researchers have identified crucial genes and pathways involved in osteosarcoma tumorigenesis, metastasis and response to therapy (Table II). This summary highlights key findings related to specific gene features, emphasizing their implications for precision medicine in osteosarcoma management.
Mutations in the TP53 gene are prevalent in numerous cancers, including osteosarcoma (56). Research indicates that TP53 missense mutations are associated with longer survival in a canine model of osteosarcoma, suggesting a potential survival advantage linked to specific mutations (57). Furthermore, the impairment of rigid sensing due to mutant TP53 gain-of-function mutations has been noted, implicating TP53 in the modulation of cellular mechanics and tumor progression (58). The myelocytomatosis viral oncogene homolog (Myc) oncogene is pivotal in osteosarcoma pathogenesis. A study demonstrated that Myc not only acts as a poor prognostic biomarker but also serves as a potential therapeutic target, highlighting its critical role in the proliferation of osteosarcoma cells (59). Furthermore, the promotion of osteosarcoma development through WW domain containing oxidoreductase-mediated upregulation of Myc adds another layer of complexity to Myc's role in this disease (60). The downregulation of GATA binding protein 3 has been linked to the epithelial-to-mesenchymal transition (EMT) and migration of osteosarcoma cells. Its regulatory effects on Slug indicate its significance in managing metastatic potential in osteosarcoma (61).
Vascular endothelial growth factor (VEGF)/VEGF receptors: VEGF is essential for angiogenesis and has been shown to be associated with poor prognosis in patients with osteosarcoma (62). The dual inhibition of VEGF and survivin has exhibited proliferation inhibition and induced apoptosis in osteosarcoma cells (63), thereby presenting a promising therapeutic strategy. In addition, the splicing factor YBX1 promotes osteosarcoma progression by upregulating VEGF and downregulating its anti-angiogenic isoform VEGF (64). Hypoxia-inducible factor 1α (HIF-1α) mediates several processes involved in osteosarcoma, including cellular response to hypoxia and metabolism. Of note, HIF-1α-mediated augmentation of certain microRNAs facilitates proliferation and metastasis, reflecting its role in EMT pathways (65). Furthermore, the interplay between HIF-1α and mTOR signaling pathways is critical, as demonstrated by the synergistic anti-tumor activity of ginsenoside Rg3 alongside doxorubicin (66).
The C-X-C motif chemokine receptor 4 (CXCR4) is crucial for osteosarcoma cell migration and metastasis (67). Studies have shown that CXCR4 interacts with mesenchymal stem cells to promote tumor growth and pulmonary metastasis, primarily through VEGF signaling (68). Additionally, the CXCR4-coactivator associated arginine methyltransferase 1 (CARM1)-Yes-associated protein 1 (YAP) axis has been implicated in overcoming doxorubicin resistance in osteosarcoma by suppressing aerobic glycolysis, underscoring potential therapeutic pathways targeting these interactions (69). Expression of matrix metalloproteinase (MMP)-2 and MMP-9 is associated with poor prognosis and higher metastatic potential in osteosarcoma (70). Furthermore, the macrophage recruitment factor monocyte chemoattractant protein-1 enhances cancer cell migration through the c-Raf/MAPK/AP-1 pathway, further indicating the role of MMPs in the metastatic cascade (71).
The integration of transcriptomic data in understanding the biology of osteosarcoma heralds a shift towards precision medicine approaches (72). Identifying specific gene expression patterns and their associated pathways allows clinicians to tailor treatment strategies based on individual molecular profiles, potentially improving outcomes. For example, targeting pathways involving TP53, HIF-1α and Myc provides a framework for developing targeted therapies that address the unique characteristics of each patient's tumor (59,73,74). Furthermore, the combination of transcriptomic and other omics data (e.g., proteomics, metabolomics) is expected to revolutionize the understanding and management of osteosarcoma. It can enable the identification of novel biomarkers for early diagnosis, prognosis and the development of innovative therapeutic strategies (75).
The application of transcriptomics in osteosarcoma research has significantly contributed to unravelling the complexities of this malignancy. Key findings regarding oncogenes, tumor suppressor genes, signaling pathways and non-coding RNAs provide a rich resource for future therapeutic strategies (76). The path toward precision medicine in osteosarcoma is being paved by these insights, which have the potential to enhance patient outcomes and transform clinical practices in oncology.
Recent advances in proteomics have illuminated the complexity of its pathology, leading to insights into specific proteins that drive cell proliferation, survival, invasion and metastasis (77). In the chapter below, key findings from proteomic studies in osteosarcoma are being discussed, focusing on significant protein classes such as cyclins, signaling proteins, apoptosis-related proteins, invasion/metastasis proteins, metabolic proteins, transcription factors and other targeted proteins (Table III).
The TME plays a crucial role in osteosarcoma progression and metastasis, largely driven by signaling pathways mediated by proteins such as RAGE, HIF-1α and CXCR4 (78). Chang et al (79) elucidate that Nε-(1-carboxymethyl)-L-lysine activates RAGE signaling, driving osteosarcoma cell metastasis via ERK/NFκB axis. This suggests a significant link between advanced glycation end products and sarcomagenesis. Overexpression of CXCR4 has been tightly correlated with disease progression and poor prognosis in osteosarcoma. Specifically, CXCR4 signaling has been shown to facilitate myeloid-derived suppressor cell accumulation, which can blunt responses to immunotherapy (80). Furthermore, CXCR4 blockade has been proven to sensitize osteosarcoma cells to doxorubicin through autophagic cell death induction and inhibition of the PI3K-Akt-mTOR pathway (81). HIF-1α, a key regulator in the tumor response to hypoxic conditions, enhances osteosarcoma proliferation and metastasis through various proteins, such as matrilin 4 (MATN4). HIF-1α mediates the expression of MATN4, promoting progression (82), indicating the dual role of hypoxic signaling in metabolic reprogramming and tumor aggressiveness.
Metastasis is a complex biological process that involves multiple signaling pathways, with several proteins acting as key facilitators. MMPs, particularly MMP-2 and MMP-9, are crucial for ECM degradation, contributing to metastasis (83). Overexpression of mucin (MUC)15 has been associated with enhanced osteosarcoma cell proliferation and invasiveness via the Wnt/β-catenin signaling pathway and regulated through MMP-2 and MMP-9 (74). A study indicated that sirtuin 6 negatively regulates proliferation and invasion in osteosarcoma by targeting N-cadherin (84), showcasing how proteins involved in EMT are leveraged by osteosarcoma cells to facilitate metastatic spread.
Proteomic studies have illuminated various pathways by which osteosarcoma cells evade programmed cell death, revealing potential targets for apoptosis-inducing therapies (77). Proteins such as survivin and Livin have been highlighted as important mediators of apoptosis resistance in osteosarcoma. MUC15 was shown to promote cell proliferation through the Livin protein, linking the Wnt/β-catenin pathway with survival (74). The B-cell lymphoma-2 (Bcl-2) inhibitor AT-101 has demonstrated efficacy in inhibiting osteosarcoma growth in preclinical models (85). Further exploration of how survival pathways modulated by Bcl-2 family members interact with proteomics will enhance our understanding of therapeutic resistance.
The metabolic profile of osteosarcoma cells is essential for their proliferation and survival. Critical proteins involved in glycolysis and oxidative phosphorylation are frequent subjects of investigation (86). A study indicated that lysine-specific demethylase 6B-mediated histone demethylation of lactate dehydrogenase A (LDHA) promotes lung metastasis in osteosarcoma. This highlights the role of LDHA in shaping the metabolic landscape (87). Furthermore, N-acetyltransferase 10-mediated ac4C acetylation of LDHA was shown to upregulate glycolytic metabolism in osteosarcoma (88). Aerobic glycolysis: The CXCR4-CARM1-YAP axis has been found to be critical in regulating glucose metabolism, linking aerobic glycolysis to chemoresistance (69). Targeting these metabolic pathways may thus provide another avenue for intervention.
Overall, the insights gleaned from proteomics research reveal numerous potential therapeutic targets for osteosarcoma. Inhibition of motif-containing family genes 26 (TRIM26) offers a potential therapeutic strategy by destabilizing RACK1 and inactivating MEK/ERK signaling, demonstrating its role in osteosarcoma progression (89). Evidence suggests that Zyxin and TRIM22 restrict proliferation and metastasis via diverse signaling pathways, providing promising avenues for therapeutic explorations (90,91). The splicing factor Y-box binding protein 1 (YBX1) has been identified as a promoter of osteosarcoma progression through the upregulation of VEGF (64). Intervening in YBX1 pathways may effectively inhibit angiogenesis and tumor growth.
The ongoing proteomics research into osteosarcoma extends our understanding of tumor biology, highlighting crucial proteins that underlie molecular pathways involved in tumor progression, metastasis and survival (92). These proteins offer promising targets for new therapeutic strategies, directing future research toward enhancing targeted therapies and personalized medicine approaches for osteosarcoma. As proteomic data are being harnessed, integrating these insights with other omics layers will be pivotal in advancing precision medicine for osteosarcoma management.
The exploration of metabolomic changes in osteosarcoma has opened up several metabolic pathways that are responsible for the disease's progression and therapeutic resistance (93). The summary provided in the chapter below delineates findings from recent studies, emphasizing various metabolic pathways related to osteosarcoma, with particular attention paid to lipid and glucose metabolism, amino acid metabolism and the role of TME interactions (Table IV).
Lipid metabolism has emerged as a critical player in osteosarcoma pathophysiology. Research indicates that deregulated lipid homeostasis is linked to cancer progression and metastasis in osteosarcoma (94). For instance, Hu et al (95) demonstrated that sphingolipid metabolism is intricately associated with osteosarcoma metastasis and patient prognosis. Furthermore, studies suggest that long non-coding RNAs, such as RPARP-antisense RNA 1, can regulate lipid metabolism, thus promoting osteosarcoma progression (96). Additionally, Bispo et al (97) have shown that novel metal-based drugs can impact lipid metabolism in MG-63 osteosarcoma cells, presenting a potential therapeutic avenue. Notably, fatty acid oxidation and the associated signaling pathways are essential for the survival and growth of osteosarcoma cells. Research by Fritsche-Guenther et al (99) highlighted the shifting nutrient dependencies of osteosarcoma cells, revealing that certain metabolic adaptations facilitate their aggressive traits. The interplay between lipid droplets and the osteosarcoma microenvironment is thus critical for understanding the tumor's metabolism.
The Warburg effect, characterized by increased glycolysis in the presence of oxygen, is prevalent among osteosarcoma cells. Shen et al (99) noted that circular RNAs, such as Hsa_circ_0000566, promote glycolysis and contribute to osteosarcoma progression by feedback regulation on HIF-1α. Similarly, Li et al (100) indicated that aberrations in glucose metabolism, specifically through the upregulation of LDHA by miR-329-3p, sensitize osteosarcoma cells to cisplatin treatment, underscoring the relevance of metabolic pathways for therapeutic responses. Furthermore, elevated glucose levels have been correlated with enhanced stemness and metastatic potential in osteosarcoma. Wang et al (101) demonstrated that modulating glucose metabolism affects the cancer stem cells' properties and their tumorigenicity. Targeting these metabolic pathways provides opportunities to enhance the efficacy of existing therapies.
Amino acid metabolism, particularly glutamine metabolism, has gained attention in cancer research due to its association with tumor growth and survival. Ren et al (102) showed that altered glutamine usage in osteosarcoma elevates drug resistance by sustaining metabolic demands under stress conditions. In addition, research by Wang et al (103) highlighted the role of YAP1-mediated glutamine metabolism in osteosarcoma, indicating that disrupting this pathway could enhance therapeutic lethality. The branched-chain amino acids (BCAAs) are also implicated in osteosarcoma progression. Lin et al (104) found that angiopoietin like 4 regulates BCAA metabolism, presenting a potential target for disrupting metabolic communications in osteosarcoma. The metabolic flexibility conferred by amino acid utilization aids osteosarcoma cells in adapting to various environmental stressors.
The TME significantly influences the metabolic landscape of osteosarcoma. A study has shown that metabolic gene expression correlates with immune microenvironment alterations, impacting clinical outcomes (105). Wu et al (106) explored the metabolic interplay between osteosarcoma cells and the immune microenvironment, suggesting that a thorough understanding of these interactions could lead to novel immunotherapy approaches. Moreover, copper metabolism, highlighted in recent work, affects both the immune landscape and the viability of osteosarcoma cells (107). Lin et al (108) emphasized the need to assess the copper-metabolism-related genes for further insights into osteosarcoma prognosis and treatment responses.
As the understanding of the metabolic alterations in osteosarcoma deepens, it brings forth exciting prospects for targeted metabolic interventions. The integration of multi-omics approaches is essential for elucidating the complex metabolic networks underlying osteosarcoma, potentially leading to personalized therapeutic strategies (93). Future research should continue to explore the interactions among different metabolic pathways and their implications in the TME, providing a foundation for innovative treatment modalities based on metabolic reprogramming.
The integration of multi-omics data provides unique challenges and opportunities. Techniques such as machine learning, bioinformatics and systems biology are becoming essential for managing the vast datasets generated by each omics layer (10). Machine learning methods, particularly those based on supervised and unsupervised algorithms, can be employed to identify patterns and correlations among genomic, transcriptomic, proteomic and metabolomic profiles. These methods facilitate the extraction of meaningful insights that a single omics approach may overlook (10). Recent advancements in computational frameworks, such as weighted gene co-expression network analysis and pathway enrichment tools, have further enabled the identification of cross-omics interactions, enhancing the interpretability of complex datasets (109).
One illustrative case study is the combined omics analysis conducted by Lin et al (110), which investigated the molecular landscape of osteosarcoma. The study utilized RNA-seq data to identify differentially expressed genes, while proteomic profiling revealed novel proteins associated with disease progression. By integrating these findings with metabolomic data, they were able to identify specific metabolic pathways, such as glycolysis and lactate metabolism, that were altered in osteosarcoma. This integrative approach provided a more holistic understanding of the TME and its metabolic adaptation, highlighting the critical interplay between different molecular layers.
A recent multi-omics study employed single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics to dissect intratumoral heterogeneity in advanced osteosarcoma (111). The study identified distinct cellular subpopulations enriched in chemotherapy-resistant cells, characterized by upregulated glycolysis and PI3K/AKT pathway activity. Proteomic validation further confirmed the overexpression of glycolytic enzymes (e.g., LDHA) and AKT1 in these subpopulations, providing a mechanistic link between metabolic reprogramming and therapeutic resistance. This study exemplifies how integrative approaches can uncover actionable targets, such as combined inhibition of AKT and glycolysis, to overcome resistance.
Another compelling example involves a study by Jia et al (112), which integrated genomic, proteomic and metabolomic data to explore cuproptosis-related pathways in osteosarcoma. The authors identified a copper metabolism signature correlated with immune suppression and poor prognosis. Functional experiments demonstrated that copper chelators synergized with immune checkpoint inhibitors to enhance T-cell infiltration and reduce tumor growth in preclinical models. This translational application underscores the potential of multi-omics in guiding combination therapies tailored to metabolic-immune crosstalk.
Furthermore, Truong et al (113) mapped the differentiation trajectories of osteosarcoma cells using scRNA-seq and chromatin accessibility assays. They revealed that epigenetic remodeling of SOX9-driven chondrogenic pathways promotes metastatic niche formation. Complementary metabolomic profiling highlighted elevated glutamine utilization in metastatic cells, suggesting therapeutic vulnerability to glutaminase inhibitors. This study not only clarified the role of SOX9 in osteosarcoma progression but also demonstrated how multi-omics can bridge molecular mechanisms to therapeutic strategies.
Integrative multi-omics approaches significantly enhance our understanding of tumor biology, yielding crucial insights that can inform treatment decisions (114). For instance, the identification of specific genetic and proteomic alterations linked to signaling pathways, such as the mTOR and PI3K/AKT pathways, allows for the development of targeted therapies aimed at these pathways (115). Wang et al (103) combined CRISPR-Cas9 screens with proteomic profiling to identify YAP1-mediated glutamine addiction in chemotherapy-resistant osteosarcoma. They demonstrated that disrupting the YAP1-glutamine axis using the small-molecule inhibitor CA3 restored cisplatin sensitivity in vivo, highlighting the translational value of pathway-centric multi-omics.
Furthermore, studies leveraging integrative approaches can identify patient stratification markers, facilitating personalized treatment regimens (116). By categorizing patients based on distinct molecular signatures, clinicians can tailor therapies that specifically target the identified vulnerabilities of each tumor. For instance, patients exhibiting specific metabolic dysregulations could be treated with agents designed to exploit those metabolic weaknesses, potentially enhancing therapeutic efficacy (117).
Furthermore, the integrative analysis of multi-omics data can assist in predicting treatment responses and resistance mechanisms in osteosarcoma (107). Understanding the interactions between genetic alterations, changes in gene expression and alterations in protein levels can provide insights into why certain patients may not respond to conventional therapies (118). For instance, the presence of specific mutations alongside corresponding protein changes can indicate an adaptive resistance mechanism, signaling the need for alternative treatment options (118).
The integrative analysis of multi-omics data can also assist in predicting treatment responses and resistance mechanisms in osteosarcoma (104). For example, Audinot et al (119) utilized circulating tumor DNA sequencing and longitudinal metabolomics to monitor osteosarcoma evolution during chemotherapy. They identified recurrent mutations in TP53 and RB1 alongside increased kynurenine pathway activity as hallmarks of acquired resistance. Targeting these pathways with poly(ADP-ribose) polymerase inhibitors and indoleamine 2,3-dioxygenase 1 blockers in patient-derived xenografts (PDXs) significantly delayed relapse, illustrating the power of dynamic multi-omics monitoring.
In summary, integrative multi-omics approaches not only provide a comprehensive view of the complex biological landscape of osteosarcoma but also foster innovation in treatment strategies. The ability to couple genomic, transcriptomic, proteomic and metabolomic data through advanced computational techniques can lead to the discovery of novel biomarkers and therapeutic targets, ultimately guiding the implementation of precision medicine in osteosarcoma management.
The integration of multi-omics approaches encompassing genomics, transcriptomics, proteomics and metabolomics holds great promise in enhancing the diagnosis and management of osteosarcoma (Table V) (4). The targets identified through multi-omics strategies show significant potential in the diagnosis and treatment of osteosarcoma, providing direction for targeted therapy and combination therapy for this condition (Fig. 2).
Mutations in TP53 have been extensively studied as potential biomarkers. Various studies indicate that TP53 mutations are associated with aggressive disease and poor outcomes in osteosarcoma (120). For instance, a meta-analysis identified TP53 mutations as significant prognostic factors, suggesting their potential as biomarkers for patient stratification (121). Specific polymorphisms in TP53 have also been linked to susceptibility to osteosarcoma among certain populations, providing insights into genetic predisposition (122). MDM2, an antagonist of TP53, has emerged as another potential biomarker. Its overexpression has been implicated in various malignancies, including osteosarcoma. Research demonstrates that MDM2 overexpression can be detected via immunohistochemistry and correlates with lower survival rates, making it a valuable adjunct in the diagnosis and prognostic assessment of patients with osteosarcoma (123). Cyclin-dependent kinases (CDKs), particularly CDK4, have garnered attention as biomarkers in osteosarcoma. Overexpression of CDK4 is associated with resistance to conventional chemotherapy and correlates with aggressive disease features (124). Furthermore, CDK4/6 inhibitors are being explored as potential therapeutic options, thus reinforcing the clinical relevance of CDK4 as a biomarker in osteosarcoma (125).
Enhancer of zeste homologue 2 (EZH2), a histone methyltransferase, has been implicated in osteosarcoma progression and poor prognosis. High levels of EZH2 expression have been associated with advanced disease stages and metastasis, establishing its role as a prognostic marker (126). Furthermore, EZH2 inhibitors are being evaluated in clinical trials, showing promise in targeted therapy approaches for osteosarcoma (127). Osteopontin has been identified as a significant serum biomarker for osteosarcoma. Elevated levels of osteopontin are associated with poorer survival outcomes, making it a potential marker for disease monitoring and therapy response (128). Its measurement in biofluids may provide real-time insights into the disease state. The use of circulating tumor DNA (ctDNA) represents a breakthrough in the non-invasive monitoring of osteosarcoma. ctDNA analysis enables the detection of tumor-specific mutations and informs about minimal residual disease and the risk of recurrence (129). Recent studies have validated the clinical utility of ctDNA quantification in predicting outcomes, indicating its integration into routine management protocols (119).
Recent advancements in metabolomics have identified various metabolites that exhibit significant alterations in patients with osteosarcoma compared to healthy controls. Specific metabolic signatures associated with tumor presence and progression may serve as novel diagnostic biomarkers, although research in this area is still in its infancy (130). Multi-omics technologies have significantly expanded the understanding of osteosarcoma, identifying various biomarkers with diagnostic and prognostic potential. Integrating these biomarkers into clinical workflows may facilitate earlier diagnosis, enhance personalized treatment strategies and ultimately improve patient outcomes. Future research should focus on validating these biomarkers in larger cohorts and elucidating their mechanisms of action, propelling the field toward precision medicine in osteosarcoma management.
VEGF has emerged as a pivotal biomarker in osteosarcoma due to its role in tumor angiogenesis. Studies have demonstrated that inhibition of VEGF not only reduces tumor growth but also enhances the sensitivity of osteosarcoma cells to therapeutic agents. For instance, silencing VEGF through microRNAs, such as miR-410, has been shown to suppress proliferation and invasion of osteosarcoma cells (131). Furthermore, the application of anti-VEGF therapies, such as bevacizumab, in combination with other agents may provide a synergistic effect, as evidenced in recent clinical trials (132,133).
The PI3K/AKT/mTOR pathway is frequently dysregulated in osteosarcoma, making it a suitable target for intervention. Various studies have explored this pathway's role in cell survival and proliferation. For instance, the natural compound rhaponticin was shown to inhibit osteosarcoma via suppression of the PI3K-Akt-mTOR signaling axis (134). Furthermore, selective inhibition of EZH2 has been demonstrated to effectively regulate the PI3K/AKT signaling pathway, impacting cancer cell behavior (135,136).
The blockade of immune checkpoints, particularly programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1), has been explored in osteosarcoma and demonstrates significant therapeutic potential. Research indicates that combining PD-1/PD-L1 inhibitors with traditional chemotherapeutics can enhance anti-tumor efficacy by overcoming immune escape mechanisms (137,138). Furthermore, the combination of immune checkpoint blockade and T-cell therapies has shown promising results in clinical settings (139).
The CDK pathway, specifically CDK4/6, has gained attention as a therapeutic target due to its involvement in cell cycle regulation in osteosarcoma (140). Targeting CDK4/6 has been shown to enhance the effects of chemotherapy and may help in the management of drug-resistant osteosarcoma variants (125,141).
In conclusion, multi-omics approaches have significantly advanced the understanding of the molecular underpinnings of osteosarcoma. By translating these findings into clinical applications, it may be possible to better harness targeted therapies, ultimately improving patient outcomes in this challenging malignancy. Continuous research and clinical trials will be paramount to validate these strategies and ensure their efficacy in the broader patient population.
One of the most pressing needs in osteosarcoma research is the establishment of collaborative research networks that promote data sharing among institutions worldwide (142). The heterogeneity of osteosarcoma, combined with the complexity of multi-omics data, necessitates a collaborative approach to facilitate large-scale studies and the development of robust databases that integrate genomic, transcriptomic, proteomic and metabolomic information (13).
Current efforts, such as the Pediatric Cancer Genome Project, provide a framework for collaborative research but need to be expanded to include international participants and diverse patient populations (143). Furthermore, data sharing platforms, such as the European Genome-phenome Archive (https://ega-archive.org) and cBioPortal (https://www.cbioportal.org), play a crucial role in providing researchers access to valuable datasets, fostering collaborations that can lead to meaningful discoveries in osteosarcoma research (144).
By encouraging multi-institutional collaborations, it may be possible to facilitate the standardization of omics data collection protocols, ensuring consistency and reproducibility among disparate studies. As multi-omics research evolves, a shared commitment to open science will enhance the overall quality of research findings and enable faster translation into clinical applications. The future of multi-omics in osteosarcoma research is closely tied to the rapid advancements in emerging technologies, particularly single-cell omics (113). Single-cell sequencing technologies allow for the analysis of individual cells within a tumor, providing unprecedented insights into tumor heterogeneity, cell lineage and microenvironment interactions that traditional bulk analysis cannot achieve (145).
These technologies hold the potential to uncover specific cell populations that drive tumor growth and metastasis, which can lead to the identification of novel biomarkers and therapeutic targets (146). Recent studies employing scRNA-seq in osteosarcoma have revealed distinct cellular subpopulations that contribute to the TME's immunosuppressive nature, potentially guiding the development of more effective immunotherapy strategies (111,147,148). Furthermore, integrating single-cell proteomics and metabolomics with transcriptomics can help create a multi-layered understanding of the molecular dynamics within tumors (111). Future initiatives should focus on standardizing these methodologies and developing comprehensive platforms that seamlessly integrate heterogeneous single-cell data for further analysis.
Longitudinal studies that utilize patient-derived samples are critical for advancing the current understanding of osteosarcoma's progression and treatment responses. Monitoring tumors over time through the collection of biopsies, blood samples for ctDNA and other biofluids can provide valuable insights into the temporal dynamics of genomic and phenotypic alterations (149). Such studies can enhance our understanding of how tumors evolve under specific therapies, allowing for the identification of resistance mechanisms and the development of adaptive treatment approaches. For instance, a prospective trial analyzing ctDNA in patients with osteosarcoma demonstrated that detecting mutations associated with chemotherapy resistance provided critical insights for treatment modifications (119).
Furthermore, incorporating PDXs and organoids derived from osteosarcoma tumors into research initiatives allows for a more accurate representation of tumor biology in the laboratory (150,151). These models can be utilized for drug screening and validation of treatment strategies before clinical implementation, ultimately promoting personalized therapy. To successfully implement these initiatives, securing funding and collaboration from stakeholders, including academic institutions, pharmaceutical companies and patient advocacy groups, will be vital. Engaging patients in the research process, ensuring their informed consent and privacy are respected, will further promote patient-centric research endeavors that align with modern ethical standards.
The future directions and initiatives in multi-omics research for osteosarcoma promise to enhance the current understanding and management of this complex disease. By fostering collaborative research and data sharing, harnessing the potential of emerging technologies such as single-cell omics, and implementing longitudinal studies using patient-derived samples, it may be possible to pave the way for precision medicine that caters to the individual needs of patients with osteosarcoma. As these initiatives evolve, they will play a vital role in overcoming the challenges currently faced by osteosarcoma research and ultimately improve patient outcomes.
In conclusion, the integration of multi-omics approaches holds transformative potential for osteosarcoma research and treatment, allowing for a comprehensive understanding of tumor heterogeneity and the development of targeted therapies. By elucidating the intricate molecular networks underlying osteosarcoma, it may be possible to enhance early diagnosis and personalize treatment strategies, ultimately improving patient outcomes. It is imperative for clinicians and researchers to collaborate and advocate for the incorporation of multi-omics into clinical practice to advance precision medicine and offer hope to patients with osteosarcoma.
Not applicable.
XC, BT, YW and XK made substantial contributions to the conception and design of the article. XC and XK were involved in the acquisition, analysis and interpretation of data. XC, BT, YW, JZ and XK wrote the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
TME |
tumor microenvironment |
|
ECM |
extracellular matrix |
|
EMT |
epithelial-mesenchymal transition |
|
VEGF |
vascular endothelial growth factor |
|
HIF-1α |
hypoxia-inducible factor 1α |
|
CXCR4 |
C-X-C chemokine receptor type 4 |
|
MMPs |
matrix metalloproteinases |
|
LDHA |
lactate dehydrogenase A |
|
YAP1 |
Yes-associated protein 1 |
|
mTOR |
mammalian target of rapamycin |
|
FGFR |
fibroblast growth factor receptor |
|
FN1 |
fibronectin 1 |
|
ICAM-1 |
intercellular adhesion molecule-1 |
|
BCAA |
branched-chain amino acid |
|
AKT1 |
AKT serine/threonine kinase 1 |
|
CLEC3A |
C-type lectin domain family 3 member A |
|
MAT1 |
ménage à trios 1 |
|
PD-1/PD-L1 |
programmed death-1/programmed death-ligand 1 |
|
CDK |
cyclin-dependent kinase |
|
EZH2 |
enhancer of zeste homologue 2 |
|
ctDNA |
circulating tumor DNA |
|
CMRG |
copper metabolism-related gene |
Not applicable.
No funding was received.
|
Ritter J and Bielack SS: Osteosarcoma. Ann Oncol. 21(Suppl 7): vii320–vii325. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Belayneh R, Fourman MS, Bhogal S and Weiss KR: Update on osteosarcoma. Curr Oncol Rep. 23:712021. View Article : Google Scholar : PubMed/NCBI | |
|
Czarnecka AM, Synoradzki K, Firlej W, Bartnik E, Sobczuk P, Fiedorowicz M, Grieb P and Rutkowski P: Molecular biology of osteosarcoma. Cancers (Basel). 12:21302020. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Y, Wang J, Sun M, Zuo D, Wang H, Shen J, Jiang W, Mu H, Ma X, Yin F, et al: Multi-omics analysis identifies osteosarcoma subtypes with distinct prognosis indicating stratified treatment. Nat Commun. 13:72072022. View Article : Google Scholar : PubMed/NCBI | |
|
Mirabello L, Troisi RJ and Savage SA: Osteosarcoma incidence and survival rates from 1973 to 2004: Data from the surveillance, epidemiology, and end results program. Cancer. 115:1531–1543. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Meltzer PS and Helman LJ: New horizons in the treatment of osteosarcoma. N Engl J Med. 385:2066–2076. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Garcia-Ortega DY, Cabrera-Nieto SA, Caro-Sánchez HS and Cruz-Ramos M: An overview of resistance to chemotherapy in osteosarcoma and future perspectives. Cancer Drug Resist. 5:762–793. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Panez-Toro I, Muñoz-García J, Vargas-Franco JW, Renodon-Cornière A, Heymann MF, Lézot F and Heymann D: Advances in osteosarcoma. Curr Osteoporos Rep. 21:330–343. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Peled Y, Levin D, Manisterski M, Kollander N, Shukrun R and Elhasid R: Weight loss and response to chemotherapy in pediatric patients with osteosarcoma. Eur J Clin Nutr. 78:541–543. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
He X, Liu X, Zuo F, Shi H and Jing J: Artificial intelligence-based multi-omics analysis fuels cancer precision medicine. Semin Cancer Biol. 88:187–200. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang L, Qu J, Harari O, Boddey JA, Wang Z and Linna-Kuosmanen S: The impact of multi-omics in medicine. Cell Rep Med. 5:1017422024. View Article : Google Scholar : PubMed/NCBI | |
|
Mohammadi-Shemirani P, Sood T and Paré G: From omics to multi-omics technologies: The discovery of novel causal mediators. Curr Atheroscler Rep. 25:55–65. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Tang S, Roberts RD, Cheng L and Li L: Osteosarcoma multi-omics landscape and subtypes. Cancers (Basel). 15:49702023. View Article : Google Scholar : PubMed/NCBI | |
|
Donisi C, Pretta A, Pusceddu V, Ziranu P, Lai E, Puzzoni M, Mariani S, Massa E, Madeddu C and Scartozzi M: Immunotherapy and cancer: The multi-omics perspective. Int J Mol Sci. 25:35632024. View Article : Google Scholar : PubMed/NCBI | |
|
Urban W, Krzystańska D, Piekarz M, Nazar J and Jankowska A: Osteosarcoma's genetic landscape painted by genes' mutations. Acta Biochim Pol. 70:671–678. 2023.PubMed/NCBI | |
|
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:djv1012015. View Article : Google Scholar : PubMed/NCBI | |
|
Mokánszki A, Chang Chien YC, Mótyán JA, Juhász P, Bádon ES, Madar L, Szegedi I, Kiss C and Méhes G: Novel RB1 and MET gene mutations in a case with bilateral retinoblastoma followed by multiple metastatic osteosarcoma. Diagnostics (Basel). 11:282020. View Article : Google Scholar : PubMed/NCBI | |
|
Righi A, Gambarotti M, Benini S, Gamberi G, Cocchi S, Picci P and Bertoni F: MDM2 and CDK4 expression in periosteal osteosarcoma. Hum Pathol. 46:549–553. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Monti P, Menichini P, Speciale A, Cutrona G, Fais F, Taiana E, Neri A, Bomben R, Gentile M, Gattei V, et al: Heterogeneity of TP53 mutations and P53 protein residual function in cancer: Does it matter? Front Oncol. 10:5933832020. View Article : Google Scholar : PubMed/NCBI | |
|
Huang MF, Wang YX, Chou YT and Lee DF: Therapeutic strategies for RB1-deficient cancers: Intersecting gene regulation and targeted therapy. Cancers (Basel). 16:15582024. View Article : Google Scholar : PubMed/NCBI | |
|
Koo N, Sharma AK and Narayan S: Therapeutics targeting p53-MDM2 interaction to induce cancer cell death. Int J Mol Sci. 23:50052022. View Article : Google Scholar : PubMed/NCBI | |
|
Synoradzki KJ, Bartnik E, Czarnecka AM, Fiedorowicz M, Firlej W, Brodziak A, Stasinska A, Rutkowski P and Grieb P: TP53 in biology and treatment of osteosarcoma. Cancers (Basel). 13:42842021. View Article : Google Scholar : PubMed/NCBI | |
|
Mannheimer JD, Tawa G, Gerhold D, Braisted J, Sayers CM, McEachron TA, Meltzer P, Mazcko C, Beck JA and LeBlanc AK: Transcriptional profiling of canine osteosarcoma identifies prognostic gene expression signatures with translational value for humans. Commun Biol. 6:8562023. View Article : Google Scholar : PubMed/NCBI | |
|
Nie JJ, Zhang B, Luo P, Luo M, Luo Y, Cao J, Wang H, Mao J, Xing Y, Liu W, et al: Enhanced pyroptosis induction with pore-forming gene delivery for osteosarcoma microenvironment reshaping. Bioact Mater. 38:455–471. 2024.PubMed/NCBI | |
|
Jin J, Cong J, Lei S, Zhang Q, Zhong X, Su Y, Lu M, Ma Y, Li Z, Wang L, et al: Cracking the code: Deciphering the role of the tumor microenvironment in osteosarcoma metastasis. Int Immunopharmacol. 121:1104222023. View Article : Google Scholar : PubMed/NCBI | |
|
Luo ZW, Liu PP, Wang ZX, Chen CY and Xie H: Macrophages in osteosarcoma immune microenvironment: Implications for immunotherapy. Front Oncol. 10:5865802020. View Article : Google Scholar : PubMed/NCBI | |
|
Ichikawa J, Schoenecker JG, Tatsuno R, Kawasaki T, Suzuki-Inoue K and Haro H: Advancing tissue factor-targeted therapy for osteosarcoma via understanding its role in the tumor microenvironment. Curr Pharm Des. 29:1009–1012. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Tatsuno R, Ichikawa J, Komohara Y, Pan C, Kawasaki T, Enomoto A, Aoki K, Hayakawa K, Iwata S, Jubashi T and Haro H: Pivotal role of IL-8 derived from the interaction between osteosarcoma and tumor-associated macrophages in osteosarcoma growth and metastasis via the FAK pathway. Cell Death Dis. 15:1082024. View Article : Google Scholar : PubMed/NCBI | |
|
Cui J, Dean D, Hornicek FJ, Chen Z and Duan Z: The role of extracelluar matrix in osteosarcoma progression and metastasis. J Exp Clin Cancer Res. 39:1782020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Y, Yang Q, Dong Y, Ji T, Zhang B, Yang C, Zheng S, Tang L, Zhou C, Qian G, et al: First-in-maintenance therapy for localized high-grade osteosarcoma: An open-label phase I/II trial of the anti-PD-L1 antibody ZKAB001. Clin Cancer Res. 29:764–774. 2023. View Article : Google Scholar | |
|
Sun Y, Zhang C, Fang Q, Zhang W and Liu W: Abnormal signal pathways and tumor heterogeneity in osteosarcoma. J Transl Med. 21:992023. View Article : Google Scholar : PubMed/NCBI | |
|
Rajan S, Franz EM, McAloney CA, Vetter TA, Cam M, Gross AC, Taslim C, Wang M, Cannon MV, Oles A and Roberts RD: Osteosarcoma tumors maintain intra-tumoral transcriptional heterogeneity during bone and lung colonization. BMC Biol. 21:982023. View Article : Google Scholar : PubMed/NCBI | |
|
Tian H, Cao J, Li B, Nice EC, Mao H, Zhang Y and Huang C: Managing the immune microenvironment of osteosarcoma: The outlook for osteosarcoma treatment. Bone Res. 11:112023. View Article : Google Scholar : PubMed/NCBI | |
|
Lilienthal I and Herold N: Targeting molecular mechanisms underlying treatment efficacy and resistance in osteosarcoma: A review of current and future strategies. Int J Mol Sci. 21:68852020. View Article : Google Scholar : PubMed/NCBI | |
|
Liao Y, Yi Q, He J, Huang D, Xiong J and Sun W and Sun W: Extracellular vesicles in tumorigenesis, metastasis, chemotherapy resistance and intercellular communication in osteosarcoma. Bioengineered. 14:113–128. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Jia C, Liu M, Yao L, Zhao F, Liu S, Li Z and Han Y: Multi-omics analysis reveals cuproptosis and mitochondria-based signature for assessing prognosis and immune landscape in osteosarcoma. Front Immunol. 14:12809452024. View Article : Google Scholar : PubMed/NCBI | |
|
Schott C, Shah AT and Sweet-Cordero EA: Genomic complexity of osteosarcoma and its implication for preclinical and clinical targeted therapies. Adv Exp Med Biol. 1258:1–19. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Li L, Sun M, Wang J and Wan S: Multi-omics based artificial intelligence for cancer research. Adv Cancer Res. 163:303–356. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Meijer DM, Ruano D, Briaire-de Bruijn IH, Wijers-Koster PM, van de Sande MAJ, Gelderblom H, Cleton-Jansen AM, de Miranda NFCC, Kuijjer ML and Bovée JVMG: The variable genomic landscape during osteosarcoma progression: Insights from a longitudinal WGS analysis. Genes Chromosomes Cancer. 63:e232532024. View Article : Google Scholar : PubMed/NCBI | |
|
Bagger FO, Borgwardt L, Jespersen AS, Hansen AR, Bertelsen B, Kodama M and Nielsen FC: Whole genome sequencing in clinical practice. BMC Med Genomics. 17:392024. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y and Yang S, Liu Y and Yang S: Deletion of Trp53 and Rb1 in Ctsk-expressing cells drives osteosarcoma progression by activating glucose metabolism and YAP signaling. MedComm (2020). 3:e1312022. View Article : Google Scholar : PubMed/NCBI | |
|
Feng W, Wang Z, Feng D, Zhu Y, Zhang K and Huang W: The effects of common variants in MDM2 and GNRH2 genes on the risk and survival of osteosarcoma in Han populations from Northwest China. Sci Rep. 10:159392020. View Article : Google Scholar : PubMed/NCBI | |
|
He X, Pang Z, Zhang X, Lan T, Chen H, Chen M, Yang H, Huang J, Chen Y, Zhang Z, et al: Consistent amplification of FRS2 and MDM2 in low-grade osteosarcoma: A genetic study of 22 cases with clinicopathologic analysis. Am J Surg Pathol. 42:1143–1155. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Limbach AL, Lingen MW, McElherne J, Mashek H, Fitzpatrick C, Hyjek E, Mostofi R and Cipriani NA: The utility of MDM2 and CDK4 immunohistochemistry and MDM2 FISH in craniofacial osteosarcoma. Head Neck Pathol. 14:889–898. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Kaur H, Kala S, Sood A, Mridha AR, Kakkar A, Yadav R, Mishra S and Mishra D: Role of MDM2, CDK4, BCL2, parafibromin and galectin 1 in differentiating osteosarcoma from its benign fibro-osseous lesions. Head Neck Pathol. 16:728–737. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ren C, Pan R, Hou L, Wu H, Sun J, Zhang W, Tian X and Chen H: Suppression of CLEC3A inhibits osteosarcoma cell proliferation and promotes their chemosensitivity through the AKT1/mTOR/HIF1α signaling pathway. Mol Med Rep. 21:1739–1748. 2020.PubMed/NCBI | |
|
Wang J, Ni J, Song D, Ding M, Huang J, Li W and He G: MAT1 facilitates the lung metastasis of osteosarcoma through upregulation of AKT1 expression. Life Sci. 234:1167712019. View Article : Google Scholar : PubMed/NCBI | |
|
Madda R, Chen CM, Chen CF, Wang JY, Wu HY, Wu PK and Chen WM: Analyzing BMP2, FGFR, and TGF beta expressions in high-grade osteosarcoma untreated and treated autografts using proteomic analysis. Int J Mol Sci. 23:74092022. View Article : Google Scholar : PubMed/NCBI | |
|
Huang YC, Chen WC, Yu CL, Chang TK, I-Chin Wei A, Chang TM, Liu JF and Wang SW: FGF2 drives osteosarcoma metastasis through activating FGFR1-4 receptor pathway-mediated ICAM-1 expression. Biochem Pharmacol. 218:1158532023. View Article : Google Scholar : PubMed/NCBI | |
|
Kim JA, Berlow NE, Lathara M, Bharathy N, Martin LR, Purohit R, Cleary MM, Liu Q, Michalek JE, Srinivasa G, et al: Sensitization of osteosarcoma to irradiation by targeting nuclear FGFR1. Biochem Biophys Res Commun. 621:101–108. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cui J, Wang W, Li Z, Zhang Z, Wu B and Zeng L: Epigenetic changes in osteosarcoma. Bull Cancer. 98:E62–E68. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Makise N, Sekimizu M, Kubo T, Wakai S, Watanabe SI, Kato T, Kinoshita T, Hiraoka N, Fukayama M, Kawai A, et al: Extraskeletal osteosarcoma: MDM2 and H3K27me3 analysis of 19 cases suggest disease heterogeneity. Histopathology. 73:147–156. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Saba KH, Cornmark L, Rissler M, Fioretos T, Åström K, Haglund F, Rosenberg AE, Brosjö O and Nord KH: Genetic profiling of a chondroblastoma-like osteosarcoma/malignant phosphaturic mesenchymal tumor of bone reveals a homozygous deletion of CDKN2A, intragenic deletion of DMD, and a targetable FN1-FGFR1 gene fusion. Genes Chromosomes Cancer. 58:731–736. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wu CC and Livingston JA: Genomics and the immune landscape of osteosarcoma. Adv Exp Med Biol. 1258:21–36. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang S, Tian Z, Feng Y, Zhang K, Pan Y, Li Y, Wang Z, Wei W, Qiao X, Zhou R, et al: Transcriptomics and metabolomics reveal changes in the regulatory mechanisms of osteosarcoma under different culture methods in vitro. BMC Med Genomics. 15:2652022. View Article : Google Scholar : PubMed/NCBI | |
|
Yoon H, Liyanarachchi S, Wright FA, Davuluri R, Lockman JC, de la Chapelle A and Pellegata NS: Gene expression profiling of isogenic cells with different TP53 gene dosage reveals numerous genes that are affected by TP53 dosage and identifies CSPG2 as a direct target of p53. Proc Natl Acad Sci USA. 99:15632–15637. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Das S, Idate R, Regan DP, Fowles JS, Lana SE, Thamm DH, Gustafson DL and Duval DL: Immune pathways and TP53 missense mutations are associated with longer survival in canine osteosarcoma. Commun Biol. 4:11782021. View Article : Google Scholar : PubMed/NCBI | |
|
Luo M, Huang M, Yang N, Zhu Y, Huang P, Xu Z, Wang W and Cai L: Impairment of rigidity sensing caused by mutant TP53 gain of function in osteosarcoma. Bone Res. 11:282023. View Article : Google Scholar : PubMed/NCBI | |
|
Feng W, Dean DC, Hornicek FJ, Spentzos D, Hoffman RM, Shi H and Duan Z: Myc is a prognostic biomarker and potential therapeutic target in osteosarcoma. Ther Adv Med Oncol. 12:17588359209220552020. View Article : Google Scholar : PubMed/NCBI | |
|
Akkawi R, Hidmi O, Haj-Yahia A, Monin J, Diment J, Drier Y, Stein GS and Aqeilan RI: WWOX promotes osteosarcoma development via upregulation of Myc. Cell Death Dis. 15:132024. View Article : Google Scholar : PubMed/NCBI | |
|
Ma L, Xue W and Ma X: GATA3 is downregulated in osteosarcoma and facilitates EMT as well as migration through regulation of slug. Onco Targets Ther. 11:7579–7589. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wu H, Zhang J, Dai R, Xu J and Feng H: Transferrin receptor-1 and VEGF are prognostic factors for osteosarcoma. J Orthop Surg Res. 14:2962019. View Article : Google Scholar : PubMed/NCBI | |
|
Gu J, Ji Z, Li D and Dong Q: Proliferation inhibition and apoptosis promotion by dual silencing of VEGF and survivin in human osteosarcoma. Acta Biochim Biophys Sin (Shanghai). 51:59–67. 2019. View Article : Google Scholar | |
|
Quan B, Li Z, Yang H, Li S, Yan X and Wang Y: The splicing factor YBX1 promotes the progression of osteosarcoma by upregulating VEGF165 and downregulating VEGF165b. Heliyon. 9:e187062023. View Article : Google Scholar | |
|
Luo P, Zhang YD, He F, Tong CJ, Liu K, Liu H, Zhu SZ, Luo JZ and Yuan B: HIF-1α-mediated augmentation of miRNA-18b-5p facilitates proliferation and metastasis in osteosarcoma through attenuation PHF2. Sci Rep. 12:103982022. View Article : Google Scholar | |
|
Zeng X, Liu S, Yang H, Jia M, Liu W and Zhu W: Synergistic anti-tumour activity of ginsenoside Rg3 and doxorubicin on proliferation, metastasis and angiogenesis in osteosarcoma by modulating mTOR/HIF-1α/VEGF and EMT signalling pathways. J Pharm Pharmacol. 75:1405–1417. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liao YX, Zhou CH, Zeng H, Zuo DQ, Wang ZY, Yin F, Hua YQ and Cai ZD: The role of the CXCL12-CXCR4/CXCR7 axis in the progression and metastasis of bone sarcomas (Review). Int J Mol Med. 32:1239–1246. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang P, Dong L, Yan K, Long H, Yang TT, Dong MQ, Zhou Y, Fan QY and Ma BA: CXCR4-mediated osteosarcoma growth and pulmonary metastasis is promoted by mesenchymal stem cells through VEGF. Oncol Rep. 30:1753–1761. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Lu H, Zhang Y, Lv J, Zhang Y, Xu T, Yang D, Duan Z, Guan Y, Jiang Z, et al: Blocking CXCR4-CARM1-YAP axis overcomes osteosarcoma doxorubicin resistance by suppressing aerobic glycolysis. Cancer Sci. 115:3305–3319. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Gong C, Sun K, Xiong HH, Sneh T, Zhang J, Zhou X, Yan P and Wang JH: Expression of CXCR4 and MMP-2 is associated with poor prognosis in patients with osteosarcoma. Histol Histopathol. 35:863–870. 2020.PubMed/NCBI | |
|
Liu JF, Chen PC, Chang TM and Hou CH: Monocyte chemoattractant protein-1 promotes cancer cell migration via c-Raf/MAPK/AP-1 pathway and MMP-9 production in osteosarcoma. J Exp Clin Cancer Res. 39:2542020. View Article : Google Scholar : PubMed/NCBI | |
|
Harrison BM and Loukopoulos P: Genomics and transcriptomics in veterinary oncology. Oncol Lett. 21:3362021. View Article : Google Scholar : PubMed/NCBI | |
|
Feleke M, Feng W, Song D, Li H, Rothzerg E, Wei Q, Kõks S, Wood D, Liu Y and Xu J: Single-cell RNA sequencing reveals differential expression of EGFL7 and VEGF in giant-cell tumor of bone and osteosarcoma. Exp Biol Med (Maywood). 247:1214–1227. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen T, Chen Z, Lian X, Wu W, Chu L, Zhang S and Wang L: MUC 15 promotes osteosarcoma cell proliferation, migration and invasion through livin, MMP-2/MMP-9 and Wnt/β-catenin signal pathway. J Cancer. 12:467–473. 2021. View Article : Google Scholar : | |
|
Li AA, Zhang Y, Li F, Zhou Y, Liu ZL and Long XH: The mechanism of VCP-mediated metastasis of osteosarcoma based on cell autophagy and the EMT pathway. Clin Transl Oncol. 25:653–661. 2023. View Article : Google Scholar | |
|
Zheng X, Liu X, Zhang X, Zhao Z, Wu W and Yu S: A single-cell and spatially resolved atlas of human osteosarcomas. J Hematol Oncol. 17:712024. View Article : Google Scholar : PubMed/NCBI | |
|
Sirikaew N, Pruksakorn D, Chaiyawat P and Chutipongtanate S: Mass spectrometric-based proteomics for biomarker discovery in osteosarcoma: Current status and future direction. Int J Mol Sci. 23:97412022. View Article : Google Scholar : PubMed/NCBI | |
|
Wu C, Gong S, Duan Y, Deng C, Kallendrusch S, Berninghausen L, Osterhoff G and Schopow N: A tumor microenvironment-based prognostic index for osteosarcoma. J Biomed Sci. 30:232023. View Article : Google Scholar : PubMed/NCBI | |
|
Chang TY, Lan KC, Wu CH, Sheu ML, Yang RS and Liu SH: Nε-(1-carboxymethyl)-L-lysine/RAGE signaling drives metastasis and cancer stemness through ERK/NFκB axis in osteosarcoma. Int J Biol Sci. 20:880–896. 2024. View Article : Google Scholar : | |
|
Jiang K, Li J, Zhang J, Wang L, Zhang Q, Ge J, Guo Y, Wang B, Huang Y, Yang T, et al: SDF-1/CXCR4 axis facilitates myeloid-derived suppressor cells accumulation in osteosarcoma microenvironment and blunts the response to anti-PD-1 therapy. Int Immunopharmacol. 75:1058182019. View Article : Google Scholar : PubMed/NCBI | |
|
Liao YX, Lv JY, Zhou ZF, Xu TY, Yang D, Gao QM, Fan L, Li GD, Yu HY and Liu KY: CXCR4 blockade sensitizes osteosarcoma to doxorubicin by inducing autophagic cell death via PI3K-Akt-mTOR pathway inhibition. Int J Oncol. 59:492021. View Article : Google Scholar : | |
|
Zhang L, Pan Y, Pan F, Huang S, Wang F, Zeng Z, Chen H and Tian X: MATN4 as a target gene of HIF-1α promotes the proliferation and metastasis of osteosarcoma. Aging (Albany NY). 16:10462–10476. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Shi X and Wang X, Yao W, Shi D, Shao X, Lu Z, Chai Y, Song J, Tang W and Wang X: Mechanism insights and therapeutic intervention of tumor metastasis: latest developments and perspectives. Signal Transduct Target Ther. 9:1922024. View Article : Google Scholar : PubMed/NCBI | |
|
Gao Y, Qu Y, Zhou Q and Ma Y: SIRT6 inhibits proliferation and invasion in osteosarcoma cells by targeting N-cadherin. Oncol Lett. 17:1237–1244. 2019.PubMed/NCBI | |
|
Masuelli L, Benvenuto M, Izzi V, Zago E, Mattera R, Cerbelli B, Potenza V, Fazi S, Ciuffa S, Tresoldi I, et al: In vivo and in vitro inhibition of osteosarcoma growth by the pan Bcl-2 inhibitor AT-101. Invest New Drugs. 38:675–689. 2020. View Article : Google Scholar | |
|
Esperança-Martins M, Fernandes I, Soares do Brito J, Macedo D, Vasques H, Serafim T, Costa L and Dias S: Sarcoma metabolomics: Current horizons and future perspectives. Cells. 10:14322021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Y, Li F, Gao B, Ma M, Chen M, Wu Y, Zhang W, Sun Y, Liu S and Shen H: KDM6B-mediated histone demethylation of LDHA promotes lung metastasis of osteosarcoma. Theranostics. 11:3868–3881. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Mei Z, Shen Z, Pu J, Liu Q, Liu G, He X, Wang Y, Yue J, Ge S, Li T, et al: NAT10 mediated ac4C acetylation driven m6A modification via involvement of YTHDC1-LDHA/PFKM regulates glycolysis and promotes osteosarcoma. Cell Commun Signal. 22:512024. View Article : Google Scholar | |
|
Xia K, Zheng D, Wei Z, Liu W and Guo W: TRIM26 inhibited osteosarcoma progression through destabilizing RACK1 and thus inactivation of MEK/ERK signaling. Cell Death Dis. 14:5292023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu W, Zhao Y, Wang G, Feng S, Ge X, Ye W, Wang Z, Zhu Y, Cai W, Bai J and Zhou X: TRIM22 inhibits osteosarcoma progression through destabilizing NRF2 and thus activation of ROS/AMPK/mTOR/autophagy signaling. Redox Biol. 53:1023442022. View Article : Google Scholar : PubMed/NCBI | |
|
Wei Z, Xia K, Zhou B, Zheng D and Guo W: Zyxin inhibits the proliferation, migration, and invasion of osteosarcoma via Rap1-mediated inhibition of the MEK/ERK signaling pathway. Biomedicines. 11:23142023. View Article : Google Scholar : PubMed/NCBI | |
|
Li G, Stampas A, Komatsu Y, Gao X, Huard J and Pan S: Proteomics in orthopedic research: Recent studies and their translational implications. J Orthop Res. 42:1631–1640. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Dean DC, Shen S, Hornicek FJ and Duan Z: From genomics to metabolomics: Emerging metastatic biomarkers in osteosarcoma. Cancer Metastasis Rev. 37:719–731. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Yin Z, Shen G, Fan M and Zheng P: Lipid metabolic reprogramming and associated ferroptosis in osteosarcoma: From molecular mechanisms to potential targets. J Bone Oncol. 51:1006602025. View Article : Google Scholar : PubMed/NCBI | |
|
Hu X, Zhou X, Zhang J and Li L: Sphingolipid metabolism is associated with osteosarcoma metastasis and prognosis: Evidence from interaction analysis. Front Endocrinol (Lausanne). 13:9836062022. View Article : Google Scholar : PubMed/NCBI | |
|
Cai F, Liu L, Bo Y, Yan W, Tao X, Peng Y, Zhang Z, Liao Q and Yi Y: LncRNA RPARP-AS1 promotes the progression of osteosarcoma cells through regulating lipid metabolism. BMC Cancer. 24:1662024. View Article : Google Scholar : PubMed/NCBI | |
|
Bispo DSC, Correia M, Carneiro TJ, Martins AS, Reis AAN, Carvalho ALMB, Marques MPM and Gil AM: Impact of conventional and potential new metal-based drugs on lipid metabolism in osteosarcoma MG-63 cells. Int J Mol Sci. 24:175562023. View Article : Google Scholar : PubMed/NCBI | |
|
Fritsche-Guenther R, Gloaguen Y, Kirchner M, Mertins P, Tunn PU and Kirwan JA: Progression-dependent altered metabolism in osteosarcoma resulting in different nutrient source dependencies. Cancers (Basel). 12:13712020. View Article : Google Scholar : PubMed/NCBI | |
|
Shen S, Xu Y, Gong Z, Yao T, Qiao D, Huang Y, Zhang Z, Gao J, Ni H, Jin Z, et al: Positive feedback regulation of circular RNA Hsa_circ_0000566 and HIF-1α promotes osteosarcoma progression and glycolysis metabolism. Aging Dis. 14:529–547. 2023.PubMed/NCBI | |
|
Li G, Li Y and Wang DY: Overexpression of miR-329-3p sensitizes osteosarcoma cells to cisplatin through suppression of glucose metabolism by targeting LDHA. Cell Biol Int. 45:766–774. 2021. View Article : Google Scholar | |
|
Wang B, Zhou Y, Zhang P, Li J and Lu X: Solasonine inhibits cancer stemness and metastasis by modulating glucose metabolism via Wnt/β-catenin/snail pathway in osteosarcoma. Am J Chin Med. 51:1293–1308. 2023. View Article : Google Scholar | |
|
Ren J, Zhao C, Sun R, Sun J, Lu L, Wu J, Li S and Cui L: Augmented drug resistance of osteosarcoma cells within decalcified bone matrix scaffold: The role of glutamine metabolism. Int J Cancer. 154:1626–1638. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Tao Y, Han J, Shen J, Mu H, Wang Z, Wang J, Jin X, Zhang Q, Yang Y, et al: Disrupting YAP1-mediated glutamine metabolism induces synthetic lethality alongside ODC1 inhibition in osteosarcoma. Cell Oncol (Dordr). 47:1845–1861. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lin S, Miao Y, Zheng X, Dong Y, Yang Q, Yang Q, Du S, Xu J, Zhou S and Yuan T: ANGPTL4 negatively regulates the progression of osteosarcoma by remodeling branched-chain amino acid metabolism. Cell Death Discov. 8:2252022. View Article : Google Scholar : PubMed/NCBI | |
|
Qian H, Lei T, Hu Y and Lei P: Expression of lipid-metabolism genes is correlated with immune microenvironment and predicts prognosis in osteosarcoma. Front Cell Dev Biol. 9:6738272021. View Article : Google Scholar : PubMed/NCBI | |
|
Wu C, Tan J, Shen H, Deng C, Kleber C, Osterhoff G and Schopow N: Exploring the relationship between metabolism and immune microenvironment in osteosarcoma based on metabolic pathways. J Biomed Sci. 31:42024. View Article : Google Scholar : PubMed/NCBI | |
|
Li Q, Fang J, Liu K, Luo P and Wang X: Multi-omic validation of the cuproptosis-sphingolipid metabolism network: Modulating the immune landscape in osteosarcoma. Front Immunol. 15:14248062024. View Article : Google Scholar : PubMed/NCBI | |
|
Lin Z, He Y, Wu Z, Yuan Y, Li X and Luo W: Comprehensive analysis of copper-metabolism-related genes about prognosis and immune microenvironment in osteosarcoma. Sci Rep. 13:150592023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen C, Wang J, Pan D, Wang X, Xu Y, Yan J, Wang L, Yang X, Yang M and Liu GP: Applications of multi-omics analysis in human diseases. MedComm (2020). 4:e3152023. View Article : Google Scholar : PubMed/NCBI | |
|
Lin Y, Yang Y, Yuan K, Yang S, Zhang S, Li H and Tang T: Multi-omics analysis based on 3D-bioprinted models innovates therapeutic target discovery of osteosarcoma. Bioact Mater. 18:459–470. 2022.PubMed/NCBI | |
|
Zhou Y, Yang D, Yang Q, Lv X, Huang W, Zhou Z, Wang Y, Zhang Z, Yuan T, Ding X, et al: Single-cell RNA landscape of intratumoral heterogeneity and immunosuppressive microenvironment in advanced osteosarcoma. Nat Commun. 11:63222020. View Article : Google Scholar : PubMed/NCBI | |
|
Jia C, Yao X, Dong Z, Wang L, Zhao F, Gao J and Cai T: Molecular landscape and prognostic value in the post-translational ubiquitination, SUMOylation and neddylation in osteosarcoma: A transcriptome study. J Inflamm Res. 17:4315–4330. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Truong DD, Weistuch C, Murgas KA, Admane P, King BL, Chauviere Lee J, Lamhamedi-Cherradi SE, Swaminathan J, Daw NC, Gordon N, et al: Mapping the single-cell differentiation landscape of osteosarcoma. Clin Cancer Res. 30:3259–3272. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Heo YJ, Hwa C, Lee GH, Park JM and An JY: Integrative multi-omics approaches in cancer research: From biological networks to clinical subtypes. Mol Cells. 44:433–443. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wei Z, Xia K, Zheng D, Gong C and Guo W: RILP inhibits tumor progression in osteosarcoma via Grb10-mediated inhibition of the PI3K/AKT/mTOR pathway. Mol Med. 29:1332023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Wen Z, Wang Q, Ren L and Zhao S: A novel stratification framework based on anoikis-related genes for predicting the prognosis in patients with osteosarcoma. Front Immunol. 14:11998692023. View Article : Google Scholar : PubMed/NCBI | |
|
Fan J, Jahed V and Klavins K: Metabolomics in bone research. Metabolites. 11:4342021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Z, Han K, Min D, Kong W, Wang S and Gao M: Identification of the methotrexate resistance-related diagnostic markers in osteosarcoma via adaptive total variation netNMF and multi-omics datasets. Front Genet. 14:12880732023. View Article : Google Scholar : PubMed/NCBI | |
|
Audinot B, Drubay D, Gaspar N, Mohr A, Cordero C, Marec-Bérard P, Lervat C, Piperno-Neumann S, Jimenez M, Mansuy L, et al: ctDNA quantification improves estimation of outcomes in patients with high-grade osteosarcoma: A translational study from the OS2006 trial. Ann Oncol. 35:559–568. 2024. View Article : Google Scholar | |
|
Schaafsma E, Takacs EM, Kaur S, Cheng C and Kurokawa M: Predicting clinical outcomes of cancer patients with a p53 deficiency gene signature. Sci Rep. 12:13172022. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Z, Guo J, Zhang K and Guo Y: TP53 mutations and survival in osteosarcoma patients: A meta-analysis of published data. Dis Markers. 2016:46395752016. View Article : Google Scholar : PubMed/NCBI | |
|
Ru JY, Cong Y, Kang WB, Yu L, Guo T and Zhao JN: Polymorphisms in TP53 are associated with risk and survival of osteosarcoma in a Chinese population. Int J Clin Exp Pathol. 8:3198–3203. 2015.PubMed/NCBI | |
|
Jeon DG, Koh JS, Cho WH, Song WS, Kong CB, Cho SH and Lee SY and Lee SY: Clinical outcome of low-grade central osteosarcoma and role of CDK4 and MDM2 immunohistochemistry as a diagnostic adjunct. J Orthop Sci. 20:529–537. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Iwata S, Tatsumi Y, Yonemoto T, Araki A, Itami M, Kamoda H, Tsukanishi T, Hagiwara Y, Kinoshita H, Ishii T, et al: CDK4 overexpression is a predictive biomarker for resistance to conventional chemotherapy in patients with osteosarcoma. Oncol Rep. 46:1352021. View Article : Google Scholar : PubMed/NCBI | |
|
Schubert NA, Chen CY, Rodríguez A, Koster J, Dowless M, Pfister SM, Shields DJ, Stancato LF, Vassal G, Caron HN, et al: Target actionability review to evaluate CDK4/6 as a therapeutic target in paediatric solid and brain tumours. Eur J Cancer. 170:196–208. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Sun R, Shen J, Gao Y, Zhou Y, Yu Z, Hornicek F, Kan Q and Duan Z: Overexpression of EZH2 is associated with the poor prognosis in osteosarcoma and function analysis indicates a therapeutic potential. Oncotarget. 7:38333–38346. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Yang X, Xu L and Yang L: Recent advances in EZH2-based dual inhibitors in the treatment of cancers. Eur J Med Chem. 256:1154612023. View Article : Google Scholar : PubMed/NCBI | |
|
Nazarizadeh A, Alizadeh-Fanalou S, Hosseini A, Mirzaei A, Salimi V, Keshipour H, Safizadeh B, Jamshidi K, Bahrabadi M and Tavakoli-Yaraki M: Evaluation of local and circulating osteopontin in malignant and benign primary bone tumors. J Bone Oncol. 29:1003772021. View Article : Google Scholar : PubMed/NCBI | |
|
Barris DM, Weiner SB, Dubin RA, Fremed M, Zhang X, Piperdi S, Zhang W, Maqbool S, Gill J, Roth M, et al: Detection of circulating tumor DNA in patients with osteosarcoma. Oncotarget. 9:12695–12704. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Kathiresan N, Selvaraj C, Pandian S, Subbaraj GK, Alothaim AS, Safi SZ and Kulathaivel L: Proteomics and genomics insights on malignant osteosarcoma. Adv Protein Chem Struct Biol. 138:275–300. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao D, Jia P, Wang W and Zhang G: VEGF-mediated suppression of cell proliferation and invasion by miR-410 in osteosarcoma. Mol Cell Biochem. 400:87–95. 2015. View Article : Google Scholar | |
|
Tawbi HA, Burgess M, Bolejack V, Van Tine BA, Schuetze SM, Hu J, D'Angelo S, Attia S, Riedel RF, Priebat DA, et al: Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): A multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 18:1493–1501. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Assi T, Watson S, Samra B, Rassy E, Le Cesne A, Italiano A and Mir O: Targeting the VEGF pathway in osteosarcoma. Cells. 10:12402021. View Article : Google Scholar : PubMed/NCBI | |
|
Mickymaray S, Alfaiz FA, Paramasivam A, Veeraraghavan VP, Periadurai ND, Surapaneni KM and Niu G: Rhaponticin suppresses osteosarcoma through the inhibition of PI3K-Akt-mTOR pathway. Saudi J Biol Sci. 28:3641–3649. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lu DG, Tang QL, Wei JH, He FY, Lu L and Tang YJ: Targeting EZH2 by microRNA-449a inhibits osteosarcoma cell proliferation, invasion and migration via regulation of PI3K/AKT signaling pathway and epithelial-mesenchymal transition. Eur Rev Med Pharmacol Sci. 24:1656–1665. 2020.PubMed/NCBI | |
|
Chen ZY, Huang HH, Li QC, Zhan FB, Wang LB, He T, Yang CH, Wang Y, Zhang Y and Quan ZX: Capsaicin reduces cancer stemness and inhibits metastasis by downregulating SOX2 and EZH2 in osteosarcoma. Am J Chin Med. 51:1041–1066. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, He S, Wu H, Xie H, Zhang T and Deng Z: Blocking the PD-1/PD-L1 axis enhanced cisplatin chemotherapy in osteosarcoma in vitro and in vivo. Environ Health Prev Med. 24:792019. View Article : Google Scholar : PubMed/NCBI | |
|
Yoshida K, Okamoto M, Sasaki J, Kuroda C, Ishida H, Ueda K, Okano S, Ideta H, Kamanaka T, Sobajima A, et al: Clinical outcome of osteosarcoma and its correlation with programmed death-ligand 1 and T cell activation markers. Onco Targets Ther. 12:2513–2518. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
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, openlabel, single-arm, phase 1-2 trial. Lancet Oncol. 21:541–550. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Y, Shen JK, Yu Z, Hornicek FJ, Kan Q and Duan Z: Expression and therapeutic implications of cyclin-dependent kinase 4 (CDK4) in osteosarcoma. Biochim Biophys Acta Mol Basis Dis. 1864:1573–1582. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Oshiro H, Tome Y, Miyake K, Higuchi T, Sugisawa N, Kanaya F, Nishida K and Hoffman RM: Combination of CDK4/6 and mTOR inhibitors suppressed doxorubicin-resistant osteosarcoma in a patient-derived orthotopic xenograft mouse model: A translatable strategy for recalcitrant disease. Anticancer Res. 41:3287–3292. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Athieniti E and Spyrou GM: A guide to multi-omics data collection and integration for translational medicine. Comput Struct Biotechnol J. 21:134–149. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Downing JR, Wilson RK, Zhang J, Mardis ER, Pui CH, Ding L, Ley TJ and Evans WE: The pediatric cancer genome project. Nat Genet. 44:619–622. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Freeberg MA, Fromont LA, D'Altri T, Romero AF, Ciges JI, Jene A, Kerry G, Moldes M, Ariosa R, Bahena S, et al: The European genome-phenome archive in 2021. Nucleic Acids Res. 50(D1): D980–D987. 2022. View Article : Google Scholar : | |
|
Jovic D, Liang X, Zeng H, Lin L, Xu F and Luo Y: Single-cell RNA sequencing technologies and applications: A brief overview. Clin Transl Med. 12:e6942022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu W, Hu H, Shao Z, Lv X, Zhang Z, Deng X, Song Q, Han Y, Guo T, Xiong L, et al: Characterizing the tumor microenvironment at the single-cell level reveals a novel immune evasion mechanism in osteosarcoma. Bone Res. 11:42023. View Article : Google Scholar : PubMed/NCBI | |
|
Huang X, Wang L, Guo H, Zhang W and Shao Z: Single-cell transcriptomics reveals the regulative roles of cancer associated fibroblasts in tumor immune microenvironment of recurrent osteosarcoma. Theranostics. 12:5877–5887. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Feng W, Dai Y, Bao M, Yuan Z, He M, Qin Z, Liao S, He J, Huang Q, et al: Single-cell transcriptomics reveals the complexity of the tumor microenvironment of treatment-naive osteosarcoma. Front Oncol. 11:7092102021. View Article : Google Scholar : PubMed/NCBI | |
|
Fu Y, Xu Y, Liu W, Zhang J, Wang F, Jian Q, Huang G, Zou C, Xie X, Kim AH, et al: Tumor-informed deep sequencing of ctDNA detects minimal residual disease and predicts relapse in osteosarcoma. EClinicalMedicine. 73:1026972024. View Article : Google Scholar : PubMed/NCBI | |
|
Landuzzi L, Manara MC, Lollini PL and Scotlandi K: Patient derived xenografts for genome-driven therapy of osteosarcoma. Cells. 10:4162021. View Article : Google Scholar : PubMed/NCBI | |
|
He A, Huang Y, Cheng W, Zhang D, He W, Bai Y, Gu C, Ma Z, He Z, Si G, et al: Organoid culture system for patient-derived lung metastatic osteosarcoma. Med Oncol. 37:1052020. View Article : Google Scholar : PubMed/NCBI |