Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.345
Filter
1.
J Nanobiotechnology ; 22(1): 151, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38575943

ABSTRACT

BACKGROUND: As the lethal bone tumor, osteosarcoma often frequently occurs in children and adolescents with locally destructive and high metastasis. Distinctive kinds of nanoplatform with high therapeutical effect and precise diagnosis for osteosarcoma are urgently required. Multimodal optical imaging and programmed treatment, including synergistic photothermal-chemodynamic therapy (PTT-CDT) elicits immunogenetic cell death (ICD) is a promising strategy that possesses high bio-imaging sensitivity for accurate osteosarcoma delineating as well as appreciable therapeutic efficacy with ignorable side-effects. METHODS AND RESULTS: In this study, mesoporous Cu and Ce based oxide nanoplatform with Arg-Gly-Asp (RGD) anchoring is designed and successfully constructed. After loading with indocyanine green, this nanoplatform can be utilized for precisely targeting and efficaciously ablating against osteosarcoma via PTT boosted CDT and the closely following ICD stimulation both in vitro and in vivo. Besides, it provides off-peak fluorescence bio-imaging in the second window of near-infrared region (NIR II, 1000-1700 nm) and Magnetic resonance signal, serves as the dual-mode contrast agents for osteosarcoma tissue discrimination. CONCLUSION: Tumor targeted Cu&Ce based mesoporous nanoplatform permits efficient osteosarcoma suppression and dual-mode bio-imaging that opens new possibility for effectively diagnosing and inhibiting the clinical malignant osteosarcoma.


Subject(s)
Bone Neoplasms , Nanoparticles , Neoplasms , Osteosarcoma , Child , Humans , Adolescent , Magnetic Resonance Imaging/methods , Magnetic Resonance Spectroscopy , Osteosarcoma/diagnostic imaging , Osteosarcoma/therapy , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/drug therapy , Immunotherapy , Cell Line, Tumor , Phototherapy
2.
Int J Mol Sci ; 25(8)2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38673726

ABSTRACT

Bone tumors, particularly osteosarcoma, are prevalent among children and adolescents. This ailment has emerged as the second most frequent cause of cancer-related mortality in adolescents. Conventional treatment methods comprise extensive surgical resection, radiotherapy, and chemotherapy. Consequently, the management of bone tumors and bone regeneration poses significant clinical challenges. Photothermal tumor therapy has attracted considerable attention owing to its minimal invasiveness and high selectivity. However, key challenges have limited its widespread clinical use. Enhancing the tumor specificity of photosensitizers through targeting or localized activation holds potential for better outcomes with fewer adverse effects. Combinations with chemotherapies or immunotherapies also present avenues for improvement. In this review, we provide an overview of the most recent strategies aimed at overcoming the limitations of photothermal therapy (PTT), along with current research directions in the context of bone tumors, including (1) target strategies, (2) photothermal therapy combined with multiple therapies (immunotherapies, chemotherapies, and chemodynamic therapies, magnetic, and photodynamic therapies), and (3) bifunctional scaffolds for photothermal therapy and bone regeneration. We delve into the pros and cons of these combination methods and explore current research focal points. Lastly, we address the challenges and prospects of photothermal combination therapy.


Subject(s)
Bone Neoplasms , Infrared Rays , Photothermal Therapy , Humans , Bone Neoplasms/therapy , Photothermal Therapy/methods , Infrared Rays/therapeutic use , Animals , Photosensitizing Agents/therapeutic use , Osteosarcoma/therapy , Osteosarcoma/pathology , Combined Modality Therapy/methods , Immunotherapy/methods , Photochemotherapy/methods , Bone Regeneration
3.
Cancer Immunol Immunother ; 73(5): 77, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38554158

ABSTRACT

The use of large animal spontaneous models of solid cancers, such as dogs with osteosarcoma (OS), can help develop new cancer immunotherapy approaches, including chimeric antigen receptor (CAR) T cells. The goal of the present study was to generate canine CAR T cells targeting the B7-H3 (CD276) co-stimulatory molecule overexpressed by several solid cancers, including OS in both humans and dogs, and to assess their ability to recognize B7-H3 expressed by canine OS cell lines or by canine tumors in xenograft models. A second objective was to determine whether a novel dual CAR that expressed a chemokine receptor together with the B7-H3 CAR improved the activity of the canine CAR T cells. Therefore, in the studies reported here we examined B7-H3 expression by canine OS tumors, evaluated target engagement by canine B7-H3 CAR T cells in vitro, and compared the relative effectiveness of B7-H3 CAR T cells versus B7-H3-CXCR2 dual CAR T cells in canine xenograft models. We found that most canine OS tumors expressed B7-H3; whereas, levels were undetectable on normal dog tissues. Both B7-H3 CAR T cells demonstrated activation and OS-specific target killing in vitro, but there was significantly greater cytokine production by B7-H3-CXCR2 CAR T cells. In canine OS xenograft models, little anti-tumor activity was generated by B7-H3 CAR T cells; whereas, B7-H3-CXCR2 CAR T cells significantly inhibited tumor growth, inducing complete tumor elimination in most treated mice. These findings indicated therefore that addition of a chemokine receptor could significantly improve the anti-tumor activity of canine B7-H3 CAR T cells, and that evaluation of this new dual CAR construct in dogs with primary or metastatic OS is warranted since such studies could provide a critical and realistic validation of the chemokine receptor concept.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Dogs , Animals , Mice , B7 Antigens/metabolism , Osteosarcoma/therapy , Bone Neoplasms/pathology , T-Lymphocytes , Receptors, Chemokine , Cell Line, Tumor
5.
JCI Insight ; 9(9)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38512420

ABSTRACT

Spinal metastases can result in severe neurologic compromise and decreased overall survival. Despite treatment advances, local disease progression is frequent, highlighting the need for novel therapies. Tumor treating fields (TTFields) impair tumor cell replication and are influenced by properties of surrounding tissue. We hypothesized that bone's dielectric properties will enhance TTFields-mediated suppression of tumor growth in spinal metastasis models. Computational modeling of TTFields intensity was performed following surgical resection of a spinal metastasis and demonstrated enhanced TTFields intensity within the resected vertebral body. Additionally, luciferase-tagged human KRIB osteosarcoma and A549 lung adenocarcinoma cell lines were cultured in demineralized bone grafts and exposed to TTFields. Following TTFields exposure, the bioluminescence imaging (BLI) signal decreased to 10%-80% of baseline, while control cultures displayed a 4.48- to 9.36-fold increase in signal. Lastly, TTFields were applied in an orthotopic murine model of spinal metastasis. After 21 days of treatment, control mice demonstrated a 5-fold increase in BLI signal compared with TTFields-treated mice. TTFields similarly prevented tumor invasion into the spinal canal and development of neurologic symptoms. Our data suggest that TTFields can be leveraged as a local therapy within minimally conductive bone of spinal metastases. This provides the groundwork for future studies investigating TTFields for patients with treatment-refractory spinal metastases.


Subject(s)
Spinal Neoplasms , Animals , Humans , Mice , Spinal Neoplasms/secondary , Spinal Neoplasms/therapy , Cell Line, Tumor , Lung Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Cell Proliferation , Disease Models, Animal , Osteosarcoma/pathology , Osteosarcoma/therapy , Female , A549 Cells , Xenograft Model Antitumor Assays
6.
Clin. transl. oncol. (Print) ; 26(3): 709-719, mar. 2024.
Article in English | IBECS | ID: ibc-230800

ABSTRACT

Purpose Primary bone and joint sarcomas of the long bone are relatively rare neoplasms with poor prognosis. An efficient clinical tool that can accurately predict patient prognosis is not available. The current study aimed to use deep learning algorithms to develop a prediction model for the prognosis of patients with long bone sarcoma. Methods Data of patients with long bone sarcoma in the extremities was collected from the Surveillance, Epidemiology, and End Results Program database from 2004 to 2014. Univariate and multivariate analyses were performed to select possible prediction features. DeepSurv, a deep learning model, was constructed for predicting cancer-specific survival rates. In addition, the classical cox proportional hazards model was established for comparison. The predictive accuracy of our models was assessed using the C-index, Integrated Brier Score, receiver operating characteristic curve, and calibration curve. Results Age, tumor extension, histological grade, tumor size, surgery, and distant metastasis were associated with cancer-specific survival in patients with long bone sarcoma. According to loss function values, our models converged successfully and effectively learned the survival data of the training cohort. Based on the C-index, area under the curve, calibration curve, and Integrated Brier Score, the deep learning model was more accurate and flexible in predicting survival rates than the cox proportional hazards model. Conclusion A deep learning model for predicting the survival probability of patients with long bone sarcoma was constructed and validated. It is more accurate and flexible in predicting prognosis than the classical CoxPH model (AU)


Subject(s)
Humans , Neoplasms, Bone Tissue/secondary , Deep Learning , Nomograms , Osteosarcoma/pathology , Osteosarcoma/therapy , Sarcoma/pathology , Extremities , Prognosis
7.
Hum Pathol ; 147: 139-147, 2024 May.
Article in English | MEDLINE | ID: mdl-38311185

ABSTRACT

Dedifferentiation traditionally is defined by descriptive criteria as a tumor showing an abrupt change in histology from a conventional, classic, low-grade appearing neoplasm to a tumor that is more cellular, pleomorphic and "high grade", with grading typically being performed by subjective criteria. The dedifferentiated areas range from areas with recognizable histologic differentiation which differs from the primary tumor (such as an osteosarcoma arising from a low-grade chondrosarcoma) to areas containing sarcomas without specific histologic differentiation (such as pleomorphic or spindle cell sarcoma). Many, but not all, dedifferentiated tumors are aggressive and associated with significantly shorter survival than their conventional counterparts, even grade 3 conventional tumors. As a result, dedifferentiated tumors are generally considered to be clinically aggressive and as a result, more aggressive surgery or the addition of (neo)adjuvant chemotherapy is often considered. However, long-term (greater than 20 year) survivors are reported in the most common dedifferentiated bone and soft tissue sarcomas. Moreover, use of mitotic criterion for defining dedifferentiation in dedifferentiated liposarcoma as well as grading (by the French system) have been found to be associated with survival. This paper reviews the literature on dedifferentiated chondrosarcoma, dedifferentiated liposarcoma, dedifferentiated chordoma and dedifferentiated parosteal osteosarcoma. As a result of that review, recommendations are advocated to identify evidence-based, objective diagnostic and grading criteria for dedifferentiation that are appropriate for each tumor type. Adding such criteria will improve consistency in diagnosis worldwide, allow easier comparison of clinical research performed on dedifferentiated tumors and help communicate (to patients and clinicians) the tumors with highest risk of clinically aggressive behavior, to allow appropriate and personalized treatment planning.


Subject(s)
Bone Neoplasms , Cell Dedifferentiation , Neoplasm Grading , Sarcoma , Soft Tissue Neoplasms , Humans , Bone Neoplasms/pathology , Sarcoma/pathology , Soft Tissue Neoplasms/pathology , Soft Tissue Neoplasms/mortality , Chondrosarcoma/pathology , Prognosis , Osteosarcoma/pathology , Osteosarcoma/mortality , Osteosarcoma/therapy , Liposarcoma/pathology
8.
Cells ; 13(4)2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38391964

ABSTRACT

Osteosarcoma (OS) is a primary bone malignancy characterized by an aggressive nature, limited treatment options, low survival rate, and poor patient prognosis. Conditionally replicative adenoviruses (CRAds) armed with immune checkpoint inhibitors hold great potential for enhanced therapeutic efficacy. The present study aims to investigate the anti-tumor efficacy of CAV2-AU-M2, a CAV2-based CRAd armed with an anti-PD-1 single-domain antibody (sdAb), against OS cell lines in vitro. The infection, conditional replication, cytopathic effects, and cytotoxicity of CAV2-AU-M2 were tested in four different OS cell lines in two-dimensional (2D) and three-dimensional (3D) cell cultures. CAV2-AU-M2 showed selective replication in the OS cells and induced efficient tumor cell lysis and death. Moreover, CAV2-AU-M2 produced an anti-PD-1 sdAb that demonstrated effective binding to the PD-1 receptors. This study demonstrated the first CRAd armed with an anti-PD-1 sdAb. This combined approach of two distinct immunotherapies is intended to enhance the anti-tumor immune response in the tumor microenvironment.


Subject(s)
Bone Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Osteosarcoma , Single-Domain Antibodies , Humans , Oncolytic Virotherapy/methods , Osteosarcoma/therapy , Bone Neoplasms/therapy , Tumor Microenvironment
9.
ACS Appl Mater Interfaces ; 16(8): 9626-9639, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38372238

ABSTRACT

The hypoxic microenvironment in osteosarcoma inevitably compromises the antitumor effect and local bone defect repair, suggesting an urgent need for sustained oxygenation in the tumor. The currently reported oxygen-releasing materials have short oxygen-releasing cycles, harmful products, and limited antitumor effects simply by improving hypoxia. Therefore, the PCL/nHA/MgO2/PDA-integrated oxygen-releasing scaffold with a good photothermal therapy effect was innovatively constructed in this work to achieve tumor cell killing and bone regeneration functions simultaneously. The material distributes MgO2 powder evenly on the scaffold material through 3D printing technology and achieves the effect of continuous oxygen release (more than 3 weeks) through its slow reaction with water. The in vitro and in vivo results also indicate that the scaffold has good biocompatibility and sustained-release oxygen properties, which can effectively induce the proliferation and osteogenic differentiation of bone mesenchymal stem cells, achieving excellent bone defect repair. At the same time, in vitro cell experiments and subcutaneous tumorigenesis experiments also confirmed that local oxygen supply can promote osteosarcoma cell apoptosis, inhibit proliferation, and reduce the expression of heat shock protein 60, thereby enhancing the photothermal therapy effect of polydopamine and efficiently eliminating osteosarcoma. Taken together, this integrated functional scaffold provides a unique and efficient approach for antitumor and tumor-based bone defect repair for osteosarcoma treatment.


Subject(s)
Bone Neoplasms , Magnesium Compounds , Osteosarcoma , Peroxides , Humans , Tissue Scaffolds , Osteogenesis , Oxygen/pharmacology , Magnesium Oxide , Bone Regeneration , Osteosarcoma/therapy , Bone Neoplasms/drug therapy , Printing, Three-Dimensional , Combined Modality Therapy , Tumor Microenvironment
10.
Int J Oncol ; 64(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38390935

ABSTRACT

Osteosarcoma (OS) is a frequently occurring primary bone tumor, mostly affecting children, adolescents and young adults. Before 1970, surgical resection was the main treatment method for OS, but the clinical results were not promising. Subsequently, the advent of chemotherapy has improved the prognosis of patients with OS. However, there is still a high incidence of metastasis or recurrence, and chemotherapy has several side effects, thus making the 5­year survival rate markedly low. Recently, chimeric antigen receptor T (CAR­T) cell therapy represents an alternative immunotherapy approach with significant potential for hematologic malignancies. Nevertheless, the application of CAR­T cells in the treatment of OS faces numerous challenges. The present review focused on the advances in the development of CAR­T cells to improve their clinical efficacy, and discussed ways to overcome the difficulties faced by CAR T­cell therapy for OS.


Subject(s)
Bone Neoplasms , Osteosarcoma , Receptors, Chimeric Antigen , Child , Humans , Adolescent , Receptors, Chimeric Antigen/genetics , Immunotherapy, Adoptive/methods , Osteosarcoma/therapy , Bone Neoplasms/therapy , T-Lymphocytes , Receptors, Antigen, T-Cell/genetics
11.
Vet Comp Oncol ; 22(2): 198-203, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38327132

ABSTRACT

Osteosarcoma is the most common malignant primary bone cancer, but it is infrequently reported in cats. Feline appendicular osteosarcoma typically exhibits good prognosis when treated with surgery alone. A retrospective multi-institutional study was conducted to identify possible prognostic factors. Cats diagnosed with appendicular osteosarcoma were included if initial staging and follow-up information were available. Data including signalment, tumour characteristics, treatment modalities, and survival outcomes were collected and analysed. Fifty-six cats were included; the femur was the most frequently affected bone. Eight cats had distant metastasis at admission and an additional 9 developed metastatic disease during follow-up, resulting in an overall metastatic rate of 30%. Forty-nine (87.5%) cats underwent surgery, and 4 also received adjuvant chemotherapy. Among operated cats, median time to local progression (TTLP), time to distant progression and tumour-specific survival (TSS) were not reached. One- and 2-year survival rates were 66% and 55%, respectively. Seven (12.5%) cats received no treatment; 1- and 2-year survival rates were 25% and 0%, respectively. Operated cats had significantly longer TTLP (P < .001) and TSS (P = .001) compared with non-operated cats. Among operated cats, young age negatively impacted local tumour progression, while the presence of distant metastasis at diagnosis was associated with a higher risk of tumour-related death. This study reaffirms the good prognosis for cats with appendicular osteosarcoma undergoing surgery, but sheds light on some additional factors to consider. Accurate initial staging is recommended, as the metastatic rate may exceed many previous estimations. Surgery substantially extends survival time, whereas the role of chemotherapy remains uncertain.


Subject(s)
Cat Diseases , Osteosarcoma , Animals , Osteosarcoma/veterinary , Osteosarcoma/therapy , Osteosarcoma/pathology , Cats , Cat Diseases/pathology , Retrospective Studies , Male , Female , Bone Neoplasms/veterinary , Bone Neoplasms/pathology , Appendiceal Neoplasms/veterinary , Appendiceal Neoplasms/pathology , Italy
12.
ACS Appl Mater Interfaces ; 16(7): 8538-8553, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38343191

ABSTRACT

Large osseous void, postsurgical neoplastic recurrence, and slow bone-cartilage repair rate raise an imperative need to develop functional scaffold in clinical osteosarcoma treatment. Herein, a bionic bilayer scaffold constituting croconaine dye-polyethylene glycol@sodium alginate hydrogel and poly(l-lactide)/hydroxyapatite polymer matrix is fabricated to simultaneously achieve a highly efficient killing of osteosarcoma and an accelerated osteochondral regeneration. First, biomimetic osteochondral structure along with adequate interfacial interaction of the bilayer scaffold provide a structural reinforcement for transverse osseointegration and osteochondral regeneration, as evidenced by upregulated specific expressions of collagen type-I, osteopontin, and runt-related transcription factor 2. Meanwhile, thermal ablation of the synthesized nanoparticles and mitochondrial dysfunction caused by continuously released hydroxyapatite induce residual tumor necrosis synergistically. To validate the capabilities of inhibiting tumor growth and promoting osteochondral regeneration of our proposed scaffold, a novel orthotopic osteosarcoma model simulating clinical treatment scenarios of bone tumors is established on rats. Based on amounts of in vitro and in vivo results, an effective killing of osteosarcoma and a suitable osteal-microenvironment modulation of such bionic bilayer composite scaffold are achieved, which provides insightful implications for photonic hyperthermia therapy against osteosarcoma and following osseous tissue regeneration.


Subject(s)
Hyperthermia, Induced , Osteosarcoma , Rats , Animals , Tissue Scaffolds/chemistry , Bionics , Biocompatible Materials/chemistry , Durapatite/chemistry , Bone Regeneration , Osteosarcoma/therapy , Tumor Microenvironment
13.
J Immunother Cancer ; 12(1)2024 01 09.
Article in English | MEDLINE | ID: mdl-38199607

ABSTRACT

BACKGROUND: The extracellular matrix (ECM) and cancer-associated fibroblasts (CAFs) play major roles in tumor progression, metastasis, and the poor response of many solid tumors to immunotherapy. CAF-targeted chimeric antigen receptor-T cell therapy cannot infiltrate ECM-rich tumors such as osteosarcoma. METHOD: In this study, we used RNA sequencing to assess whether the recently invented membrane-anchored and tumor-targeted IL-12-armed (attIL12) T cells, which bind cell-surface vimentin (CSV) on tumor cells, could destroy CAFs to disrupt the ECM. We established an in vitro model of the interaction between osteosarcoma CAFs and attIL12-T cells to uncover the underlying mechanism by which attIL12-T cells penetrate stroma-enriched osteosarcoma tumors. RESULTS: RNA sequencing demonstrated that attIL12-T cell treatment altered ECM-related gene expression. Immunohistochemistry staining revealed disruption or elimination of high-density CAFs and ECM in osteosarcoma xenograft tumors following attIL12-T cell treatment, and CAF/ECM density was inversely correlated with T-cell infiltration. Other IL12-armed T cells, such as wild-type IL-12-targeted or tumor-targeted IL-12-T cells, did not disrupt the ECM because this effect depended on the engagement between CSV on the tumor cell and its ligand on the attIL12-T cells. Mechanistic studies found that attIL12-T cell treatment elevated IFNγ production on interacting with CSV+ tumor cells, suppressing transforming growth factor beta secretion and in turn upregulating FAS-mediated CAF apoptosis. CAF destruction reshaped the tumor stroma to favor T-cell infiltration and tumor inhibition. CONCLUSIONS: This study unveiled a novel therapy-attIL12-T cells-for targeting CAFs/ECM. These findings are highly relevant to humans because CAFs are abundant in human osteosarcoma.


Subject(s)
Bone Neoplasms , Cancer-Associated Fibroblasts , Osteosarcoma , Animals , Humans , Interleukin-12 , Heterografts , Osteosarcoma/therapy , Cell Membrane , Extracellular Matrix , Disease Models, Animal , Bone Neoplasms/therapy , Cell- and Tissue-Based Therapy
14.
Eur J Med Res ; 29(1): 45, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38212774

ABSTRACT

There are more than 170 subtypes of sarcomas (SARC), which pose a challenge for diagnosis and patient management. Relatively simple or complex karyotypes play an indispensable role in the early diagnosis and effective treatment of SARC. The genes related to absorption, distribution, metabolism, and excretion (ADME) of a drug can serve as prognostic biomarkers of cancer and potential drug targets. In this study, a risk score signature was created. The SARC cohort was downloaded from The Cancer Genome Atlas (TCGA) database, and divided into high-risk group and low-risk group according to the median value of risk score. Compared with high-risk group, low-risk group has a longer survival time, which is also verified in osteosarcoma cohort from Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database. In addition, the relationship between the signature and immunophenotypes, including status of immune cell infiltration and immune checkpoint expression, was explored. Then, we found that high-risk group is in immunosuppressive status. Finally, we verified that PPARD played a role as a carcinogen in osteosarcoma, which provided a direction for targeted treatment of osteosarcoma in the future. Generally speaking, the signature can not only help clinicians predict the prognosis of patients with SARC, but also provide a theoretical basis for developing more effective targeted drugs in the future.


Subject(s)
Bone Neoplasms , Osteosarcoma , Sarcoma , Soft Tissue Neoplasms , Humans , Prognosis , Sarcoma/genetics , Sarcoma/therapy , Osteosarcoma/genetics , Osteosarcoma/therapy , Immunotherapy
15.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(2): 197-203, 2024 Feb 09.
Article in Chinese | MEDLINE | ID: mdl-38280741

ABSTRACT

Osteosarcoma of the jaw (JOS), is a relatively rare type of osteosarcoma, with a unique pathogenesis and pathological manifestations. The clinical manifestation of JOS is not characteristic, and it often needs to be diagnosed by combining radiological and pathological examination. At present, the conventional treatment of JOS is a comprehensive treatment based on surgery and supplemented by radiotherapy and chemotherapy. Recently, the emergence of new therapies such as immunotherapy, gene therapy, phototherapy and traditional Chinese medicine has provided more choices for treatment and brought new hope to patients with JOS. Therefore, this article summarized the current understanding of diagnosis and the latest treatment development of JOS.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Nigeria , Osteosarcoma/therapy , Osteosarcoma/drug therapy , Bone Neoplasms/diagnosis , Bone Neoplasms/therapy
16.
J Neurooncol ; 166(2): 293-301, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38225469

ABSTRACT

PURPOSE: Primary osseous neoplasms of the spine, including Ewing's sarcoma, osteosarcoma, chondrosarcoma, and chordoma, are rare tumors with significant morbidity and mortality. The present study aims to identify the prevalence and impact of racial disparities on management and outcomes of patients with these malignancies. METHODS: The 2000 to 2020 Surveillance, Epidemiology, and End Results (SEER) Registry, a cancer registry, was retrospectively reviewed to identify patients with Ewing's sarcoma, osteosarcoma, chondrosarcoma, or chordoma of the vertebral column or sacrum/pelvis. Study patients were divided into race-based cohorts: White, Black, Hispanic, and Other. Demographics, tumor characteristics, treatment variables, and mortality were assessed. RESULTS: 2,415 patients were identified, of which 69.8% were White, 5.8% Black, 16.1% Hispanic, and 8.4% classified as "Other". Tumor type varied significantly between cohorts, with osteosarcoma affecting a greater proportion of Black patients compared to the others (p < 0.001). A lower proportion of Black and Other race patients received surgery compared to White and Hispanic patients (p < 0.001). Utilization of chemotherapy was highest in the Hispanic cohort (p < 0.001), though use of radiotherapy was similar across cohorts (p = 0.123). Five-year survival (p < 0.001) and median survival were greatest in White patients (p < 0.001). Compared to non-Hispanic Whites, Hispanic (p < 0.001) and "Other" patients (p < 0.001) were associated with reduced survival. CONCLUSION: Race may be associated with tumor characteristics at diagnosis (including subtype, size, and site), treatment utilization, and mortality, with non-White patients having lower survival compared to White patients. Further studies are necessary to identify underlying causes of these disparities and solutions for eliminating them.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Chordoma , Osteosarcoma , Sarcoma, Ewing , Humans , Sarcoma, Ewing/pathology , Sarcoma, Ewing/surgery , Chordoma/pathology , Retrospective Studies , SEER Program , Osteosarcoma/therapy , Chondrosarcoma/pathology , Spine/pathology , Bone Neoplasms/therapy
17.
Pediatr Dev Pathol ; 27(1): 32-38, 2024.
Article in English | MEDLINE | ID: mdl-37943723

ABSTRACT

INTRODUCTION: In osteosarcoma, the most significant indicator of prognosis is the histologic changes related to tumor response to preoperative chemotherapy, such as necrosis. We have developed a method to measure the osteosarcoma treatment effect using whole slide image (WSI) with an open-source digital image analytical software Qupath. MATERIALS AND METHODS: In Qupath, each osteosarcoma case was treated as a project. All H&E slides from the entire representative slice of osteosarcoma were scanned into WSIs and imported into a project in Qupath. The regions of tumor and tumor necrosis were annotated, and their areas were measured in Qupath. In order to measure the osteosarcoma treatment effect, we needed to calculate the percentage of total necrosis area over total tumor area. We developed a tool that can automatically extract all values of tumor and necrosis areas from a Qupath project into an Excel file, sum these values for necrosis and whole tumor respectively, and calculate necrosis/tumor percentage. CONCLUSION: Our method that combines WSI with Qupath can provide an objective measurement to facilitate pathologist's assessment of osteosarcoma response to treatment. The proposed approach can also be used for other types of tumors that have clinical need for post-treatment response assessment.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Software , Osteosarcoma/diagnosis , Osteosarcoma/therapy , Osteosarcoma/pathology , Bone Neoplasms/diagnosis , Bone Neoplasms/therapy , Bone Neoplasms/pathology , Necrosis/pathology
18.
J Gene Med ; 26(1): e3572, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37525871

ABSTRACT

BACKGROUND: The physiological and immunological characteristics of the tumor microenvironment (TME) have a profound impact on the effectiveness of immunotherapy. The present study aimed to define the TME subtype of osteosarcoma according to the signatures representing the global TME of the tumor, as well as create a new prognostic assessment tool to monitor the prognosis, TME activity and immunotherapy response of patients with osteosarcoma. METHODS: The enrichment scores of 29 functional gene expression signatures in osteosarcoma samples were calculated by single sample gene set enrichment analysis (ssGSEA). TME classification of osteosarcoma was performed and a prognostic assessment tool was created based on 29 ssGSEA scores to comprehensively correlate them with TME components, immunotherapy efficacy and prognosis of osteosarcoma. RESULTS: Three TME subtypes were generated that differed in survival, TME activity and immunotherapeutic response. Four differentially expressed genes between TME subtypes were involved in the development of prognostic assessment tools. The established prognosis assessment tool had strong performance in both training and verification cohorts, could be effectively applied to the survival prediction of samples of different ages, genders and transfer states, and could well distinguish the TME status of different samples. CONCLUSIONS: The present study describes three different TME phenotypes in osteosarcoma, provides a risk stratification tool for osteosarcoma prognosis and TME status assessment, and provides additional information for clinical decision-making of immunotherapy.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Female , Male , Prognosis , Tumor Microenvironment/genetics , Osteosarcoma/diagnosis , Osteosarcoma/genetics , Osteosarcoma/therapy , Phenotype , Immunotherapy , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Bone Neoplasms/therapy
19.
Clin Transl Oncol ; 26(3): 709-719, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37552409

ABSTRACT

PURPOSE: Primary bone and joint sarcomas of the long bone are relatively rare neoplasms with poor prognosis. An efficient clinical tool that can accurately predict patient prognosis is not available. The current study aimed to use deep learning algorithms to develop a prediction model for the prognosis of patients with long bone sarcoma. METHODS: Data of patients with long bone sarcoma in the extremities was collected from the Surveillance, Epidemiology, and End Results Program database from 2004 to 2014. Univariate and multivariate analyses were performed to select possible prediction features. DeepSurv, a deep learning model, was constructed for predicting cancer-specific survival rates. In addition, the classical cox proportional hazards model was established for comparison. The predictive accuracy of our models was assessed using the C-index, Integrated Brier Score, receiver operating characteristic curve, and calibration curve. RESULTS: Age, tumor extension, histological grade, tumor size, surgery, and distant metastasis were associated with cancer-specific survival in patients with long bone sarcoma. According to loss function values, our models converged successfully and effectively learned the survival data of the training cohort. Based on the C-index, area under the curve, calibration curve, and Integrated Brier Score, the deep learning model was more accurate and flexible in predicting survival rates than the cox proportional hazards model. CONCLUSION: A deep learning model for predicting the survival probability of patients with long bone sarcoma was constructed and validated. It is more accurate and flexible in predicting prognosis than the classical CoxPH model.


Subject(s)
Bone Neoplasms , Deep Learning , Osteosarcoma , Sarcoma , Humans , Sarcoma/pathology , Bone Neoplasms/secondary , Prognosis , Osteosarcoma/therapy , Osteosarcoma/pathology , Extremities/pathology , Nomograms
20.
Curr Cancer Drug Targets ; 24(3): 308-318, 2024.
Article in English | MEDLINE | ID: mdl-37581517

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is the most common primary malignant tumor of bone tissue, which has an insidious onset and is difficult to detect early, and few early diagnostic markers with high specificity and sensitivity. Therefore, this study aims to identify potential biomarkers that can help diagnose OS in its early stages and improve the prognosis of patients. METHODS: The data sets of GSE12789, GSE28424, GSE33382 and GSE36001 were combined and normalized to identify Differentially Expressed Genes (DEGs). The data were analyzed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genome (KEGG) and Disease Ontology (DO). The hub gene was selected based on the common DEG that was obtained by applying two regression methods: the Least Absolute Shrinkage and Selection Operator (LASSO) and Support vVector Machine (SVM). Then the diagnostic value of the hub gene was evaluated in the GSE42572 data set. Finally, the correlation between immunocyte infiltration and key genes was analyzed by CIBERSORT. RESULTS: The regression analysis results of LASSO and SVM are the following three DEGs: FK501 binding protein 51 (FKBP5), C-C motif chemokine ligand 5 (CCL5), complement component 1 Q subcomponent B chain (C1QB). We evaluated the diagnostic performance of three biomarkers (FKBP5, CCL5 and C1QB) for osteosarcoma using receiver operating characteristic (ROC) analysis. In the training group, the area under the curve (AUC) of FKBP5, CCL5 and C1QB was 0.907, 0.874 and 0.676, respectively. In the validation group, the AUC of FKBP5, CCL5 and C1QB was 0.618, 0.932 and 0.895, respectively. It is noteworthy that these genes were more expressed in tumor tissues than in normal tissues by various immune cell types, such as plasma cells, CD8+ T cells, T regulatory cells (Tregs), activated NK cells, activated dendritic cells and activated mast cells. These immune cell types are also associated with the expression levels of the three diagnostic genes that we identified. CONCLUSION: We found that CCL5 can be considered an early diagnostic gene of osteosarcoma, and CCL5 interacts with immune cells to influence tumor occurrence and development. These findings have important implications for the early detection of osteosarcoma and the identification of novel therapeutic targets.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Ligands , Osteosarcoma/diagnosis , Osteosarcoma/genetics , Osteosarcoma/therapy , Biomarkers , Immunotherapy , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Bone Neoplasms/therapy , Chemokine CCL5/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...