Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.149
Filter
1.
Biomaterials ; 312: 122714, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39079462

ABSTRACT

Osteosarcoma, a malignant bone tumor often characterized by high hedgehog signaling activity, residual tumor cells, and substantial bone defects, poses significant challenges to both treatment response and postsurgical recovery. Here, we developed a nanocomposite hydrogel for the sustained co-delivery of bioactive magnesium ions, anti-PD-L1 antibody (αPD-L1), and hedgehog pathway antagonist vismodegib, to eradicate residual tumor cells while promoting bone regeneration post-surgery. In a mouse model of tibia osteosarcoma, this hydrogel-mediated combination therapy led to remarkable tumor growth inhibition and hence increased animal survival by enhancing the activity of tumor-suppressed CD8+ T cells. Meanwhile, the implanted hydrogel improved the microenvironment of osteogenesis through long-term sustained release of Mg2+, facilitating bone defect repair by upregulating the expression of osteogenic genes. After 21 days, the expression levels of ALP, COL1, RUNX2, and BGLAP in the Vis-αPD-L1-Gel group were approximately 4.1, 5.1, 5.5, and 3.4 times higher than those of the control, respectively. We believe that this hydrogel-based combination therapy offers a potentially valuable strategy for treating osteosarcoma and addressing the tumor-related complex bone diseases.


Subject(s)
Bone Neoplasms , Hydrogels , Immunotherapy , Nanocomposites , Osteosarcoma , Osteosarcoma/pathology , Osteosarcoma/drug therapy , Osteosarcoma/therapy , Animals , Hydrogels/chemistry , Nanocomposites/chemistry , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Mice , Immunotherapy/methods , Cell Line, Tumor , Bone Regeneration/drug effects , Humans , Osteogenesis/drug effects , B7-H1 Antigen/metabolism , Mice, Inbred BALB C , Magnesium/chemistry
2.
Biomaterials ; 312: 122724, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39106818

ABSTRACT

The residual bone tumor and defects which is caused by surgical therapy of bone tumor is a major and important problem in clinicals. And the sequential treatment for irradiating residual tumor and repairing bone defects has wildly prospects. In this study, we developed a general modification strategy by gallic acid (GA)-assisted coordination chemistry to prepare black calcium-based materials, which combines the sequential photothermal therapy of bone tumor and bone defects. The GA modification endows the materials remarkable photothermal properties. Under the near-infrared (NIR) irradiation with different power densities, the black GA-modified bone matrix (GBM) did not merely display an excellent performance in eliminating bone tumor with high temperature, but showed a facile effect of the mild-heat stimulation to accelerate bone regeneration. GBM can efficiently regulate the microenvironments of bone regeneration in a spatial-temporal manner, including inflammation/immune response, vascularization and osteogenic differentiation. Meanwhile, the integrin/PI3K/Akt signaling pathway of bone marrow mesenchymal stem cells (BMSCs) was revealed to be involved in the effect of osteogenesis induced by the mild-heat stimulation. The outcome of this study not only provides a serial of new multifunctional biomaterials, but also demonstrates a general strategy for designing novel blacked calcium-based biomaterials with great potential for clinical use.


Subject(s)
Bone Neoplasms , Bone Regeneration , Calcium , Gallic Acid , Mesenchymal Stem Cells , Gallic Acid/chemistry , Bone Regeneration/drug effects , Animals , Calcium/metabolism , Bone Neoplasms/therapy , Bone Neoplasms/drug therapy , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Photothermal Therapy/methods , Osteogenesis/drug effects , Mice , Humans , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Line, Tumor
3.
Cells ; 13(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39273017

ABSTRACT

PAK4 and PD-L1 have been suggested as novel therapeutic targets in human cancers. Moreover, PAK4 has been suggested to be a molecule closely related to the immune evasion of cancers. Therefore, this study evaluated the roles of PAK4 and PD-L1 in the progression of osteosarcomas in 32 osteosarcomas and osteosarcoma cells. In human osteosarcomas, immunohistochemical positivity for the expression of PAK4 (overall survival, p = 0.028) and PD-L1 (relapse-free survival, p = 0.002) were independent indicators for the survival of patients in a multivariate analysis. In osteosarcoma cells, the overexpression of PAK4 increased proliferation and invasiveness, while the knockdown of PAK4 suppressed proliferation and invasiveness. The expression of PAK4 was associated with the expression of the molecules related to cell cycle regulation, invasion, and apoptosis. PAK4 was involved in resistance to apoptosis under a treatment regime with doxorubicin for osteosarcoma. In U2OS cells, PAK4 was involved in the stabilization of PD-L1 from ubiquitin-mediated proteasomal degradation and the in vivo infiltration of immune cells such as regulatory T cells and PD1-, CD4-, and CD8-positive cells in mice tumors. In conclusion, this study suggests that PAK4 is involved in the progression of osteosarcoma by promoting proliferation, invasion, and resistance to doxorubicin and stabilized PD-L1 from proteasomal degradation.


Subject(s)
B7-H1 Antigen , Cell Proliferation , Doxorubicin , Drug Resistance, Neoplasm , Osteosarcoma , p21-Activated Kinases , Osteosarcoma/pathology , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/genetics , Humans , B7-H1 Antigen/metabolism , Female , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Animals , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Male , Cell Line, Tumor , Cell Proliferation/drug effects , Mice , Apoptosis/drug effects , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Adult , Adolescent , Protein Stability/drug effects , Mice, Nude , Young Adult , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Invasiveness
4.
Medicine (Baltimore) ; 103(36): e39546, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39252261

ABSTRACT

Ewing sarcoma (ES) is a small round cell malignancy, mainly in the bone tissue, followed by the soft tissue. Lung metastases (LM) and bone metastases (BM) are the most common types of metastases. From 2010 to 2018, the Surveillance, Epidemiology, and End Results database diagnosed 242 cases of ES with LM, 186 cases of ES with BM, and 74 cases of ES with LM and BM. Univariate and multivariate logistic regression analyses were used to determine the risk factors for LM and/or BM, and Kaplan-Meier curves and Cox regression analysis were used to determine the prognostic factors for LM and/or BM. Tumor size ≥50 mm, N1 stage, BM, liver metastases, and surgical treatment were significantly correlated with LM; tumor size >100 mm, brain metastases, LM, surgical treatment, and chemotherapy were significantly correlated with BM; female, N1 stage, brain metastases, liver metastases, and surgical treatment were significantly correlated with LM and BM. Older age, BM, higher T stage, no surgical treatment, and no chemotherapy were harmful to the survival of ES patients with LM; older age, female, LM, and no chemotherapy were harmful to the survival of ES patients with BM; older age and no chemotherapy were harmful to the survival of ES patients with LM and BM. Larger tumor size, N1 stages, and organ metastases were significantly associated with ES patients with LM and/or BM. Chemotherapy is effective in improving the survival.


Subject(s)
Bone Neoplasms , Lung Neoplasms , Sarcoma, Ewing , Humans , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/mortality , Sarcoma, Ewing/pathology , Bone Neoplasms/secondary , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Male , Female , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lung Neoplasms/mortality , Adult , Adolescent , Young Adult , SEER Program , Child , Middle Aged , Kaplan-Meier Estimate , Prognosis , Risk Factors , Brain Neoplasms/secondary , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Retrospective Studies , Neoplasm Staging , Child, Preschool
5.
Biomed Mater ; 19(6)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39226916

ABSTRACT

Osteosarcoma (OS) is the mostly commonly occurring primary bone cancer. Despite comprehensive treatment programs including neoadjuvant chemotherapy and tumour resection, survival rates have not improved significantly since the 1970s. Survival rates are dramatically reduced for patients who suffer a local recurrence. Furthermore, primary bone cancer patients are at increased risk of bone fractures. Consequently, there is an urgent need for alternative treatment options. In this paper we report the development of novel gallium doped bioactive glass that selectively kill bone cancer cells whilst simultaneously stimulating new bone growth. Here we show, using a combination of 3-(4.5-dimethylthiazol-2-yl)-2.5-diphenyltetrazolium bromide, LIVE/DEAD assays and image analysis, that bioactive glasses containing gallium oxide are highly toxic and reduce both the proliferation and migration of bone cancer cells (Saos-2) in a dose dependant manner. Glasses containing 5 mol% gallium oxide reduced the viability of OS cells by 99% without being cytotoxic to the non-cancerous normal human osteoblasts (NHOst) control cells. Furthermore, Fourier transform infrared and energy-dispersive x-ray spectroscopy results confirmed the formation of an amorphous calcium phosphate/hydroxyapatite like layer on the surface of the bioactive glass particulates, after 7 d incubating in simulated body fluid, indicating the early stages of bone formation. These materials show significant potential for use in bone cancer applications as part of a multimodal treatment.


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Cell Proliferation , Cell Survival , Gallium , Glass , Osteosarcoma , Humans , Gallium/chemistry , Osteosarcoma/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Glass/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cell Proliferation/drug effects , Bone Neoplasms/drug therapy , Spectroscopy, Fourier Transform Infrared , Osteoblasts/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Movement/drug effects , Drug Delivery Systems , Materials Testing
6.
Cancer Med ; 13(18): e70239, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39300969

ABSTRACT

BACKGROUND: Osteosarcoma (OS) survival rates and outcome have not improved in 50 years since the advent of modern chemotherapeutics. Thus, there is a critical need for an improved understanding of the tumor microenvironment to identify better therapies. Extracellular matrix (ECM) deposition and hypoxia are known to abrogate the efficacy of various chemical and cell-based therapeutics. Here, we aim to mechanistically investigate the combinatorial effects of hypoxia and matrix deposition with the use of OS spheroids. METHODS: We use two murine OS cell lines with differential metastatic potential to form spheroids. We form spheroids of two sizes, use ascorbate-2-phosphate supplementation to enhance ECM deposition, and study cell response under standard (21% O2) and physiologic (5% O2) oxygen tensions. Finally, we examine chemotherapeutic responses to doxorubicin treatment. RESULTS: ECM production and oxygen tension are key determinants of spheroid size through cell organization based on nutrient and oxygen distribution. Interestingly, highly metastatic OS is more susceptible to chemotherapeutics compared to less metastatic OS when matrix production increases. Together, these data suggest that dynamic interactions between ECM production and oxygen diffusion may result in distinct chemotherapeutic responses despite inherent tumor aggressiveness. CONCLUSION: This work establishes OS spheroids as a valuable tool for early OS tumor formation investigation and holds potential for novel therapeutic target and prognostic indicator discovery.


Subject(s)
Extracellular Matrix , Osteosarcoma , Oxygen , Spheroids, Cellular , Tumor Microenvironment , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/pathology , Spheroids, Cellular/drug effects , Extracellular Matrix/metabolism , Animals , Mice , Oxygen/metabolism , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
7.
Int J Oncol ; 65(5)2024 11.
Article in English | MEDLINE | ID: mdl-39301646

ABSTRACT

The incidence of prostate cancer (PCa) is increasing, making it one of the prevalent malignancies among men. Metastasis of PCa to the bones poses the greatest danger to patients, potentially resulting in treatment ineffectiveness and mortality. At present, the management of patients with bone metastasis focuses primarily on providing palliative care. Research has indicated that the spread of PCa to the bones occurs through the participation of numerous molecules and their respective pathways. Gaining knowledge regarding the molecular processes involved in bone metastasis may result in the development of innovative and well­tolerated therapies, ultimately enhancing the quality of life and prognosis of patients. The present article provides the latest overview of the molecular mechanisms involved in the formation of bone metastatic tumors from PCa. Additionally, the clinical outcomes of targeted drug therapies for bone metastasis are thoroughly analyzed. Finally, the benefits and difficulties of targeted therapy for bone metastasis of PCa are discussed, aiming to offer fresh perspectives for treatment.


Subject(s)
Bone Neoplasms , Molecular Targeted Therapy , Prostatic Neoplasms , Humans , Bone Neoplasms/secondary , Bone Neoplasms/therapy , Bone Neoplasms/drug therapy , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/therapy , Molecular Targeted Therapy/methods , Antineoplastic Agents/therapeutic use , Quality of Life
8.
Cancer Med ; 13(18): e70206, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39324173

ABSTRACT

BACKGROUND: The poor overall survival of osteosarcoma (OS) underscores the need to explore new therapeutic avenues. Tumor necrosis rate (TNR) after neoadjuvant chemotherapy predicts prognosis. AIMS: The study was to investigate safety and activity of neoadjuvant chemotherapy with camrelizumab (a humanized antibody against PD-1) in patients with resectable OS. MATERIALS & METHODS: We conducted a prospective, single-arm, exploratory phase II trial in OS patients. Eligible patients received camrelizumab combined with doxorubicin or liposomal doxorubicin, cisplatin, methotrexate, ifosfamide with mesna. Surgery was performed 12-14 days after neoadjuvant therapy and adjuvant therapy starting 2-3 weeks postoperatively. The primary endpoint was the rate of good tumor necrosis (TNR ≥90%) after neoadjuvant therapy, and the secondary outcomes were safety, 2-year progression free survival and 2-year overall survival. RESULTS: Seventy-five patients were recruited to the study. Subsequently, 64 patients completed neoadjuvant therapy and underwent surgery. Thirty-one patients (48.4%) have a good TNR to neoadjuvant therapy. With a median follow-up of 22.4 months (range 2.2-44.9 months), the estimated 2-year PFS was 69.6% and the estimated 2-year overall survival was 89.4%. Grade 3 or 4 treatment-related adverse events were noticed in 62.7% of the patients. Frequent grade 3 or 4 adverse events were decreased platelet count (45.3%), decreased white blood cell count (36%). No immune-related serious adverse events were observed. DISCUSSION: Our study had limitations. First, it was limited by its non-randomized design. Besides, stromal tumor-infiltrating lymphocytes was comprehensively analyzed in this study. CONCLUSIONS: This study demonstrated that amrelizumab combined with adriamycin, cisplatin, methotrexate, and ifosfamide in the neoadjuvant treatment of resectable OS was safe and tolerable. This combined therapeutic strategy may not increase TNR, but the long-term survival benefit remains to be followed up.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Cisplatin , Doxorubicin , Ifosfamide , Methotrexate , Neoadjuvant Therapy , Osteosarcoma , Humans , Female , Osteosarcoma/drug therapy , Osteosarcoma/mortality , Osteosarcoma/therapy , Osteosarcoma/surgery , Male , Methotrexate/administration & dosage , Methotrexate/therapeutic use , Ifosfamide/administration & dosage , Ifosfamide/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Neoadjuvant Therapy/methods , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Adult , Prospective Studies , Young Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Adolescent , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Middle Aged
9.
Cell Rep Med ; 5(9): 101728, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39293390

ABSTRACT

Despite advances in treatment, the prognosis of patients with osteosarcoma remains unsatisfactory, and searching for potential targets is imperative. Here, we identify N4-acetylcytidine (ac4C) acetyltransferase 10 (NAT10) as a candidate therapeutic target in osteosarcoma through functional screening. NAT10 overexpression is correlated with a poor prognosis, and NAT10 knockout inhibits osteosarcoma progression. Mechanistically, NAT10 enhances mRNA stability of activating transcription factor 4 (ATF4) through ac4C modification. ATF4 induces the transcription of asparagine synthetase (ASNS), which catalyzes asparagine (Asn) biosynthesis, facilitating osteosarcoma progression. Utilizing virtual screening, we identify paliperidone and AG-401 as potential NAT10 inhibitors, and both inhibitors are found to bind to NAT10 proteins. Inhibiting NAT10 suppresses osteosarcoma progression in vivo. Combined treatment using paliperidone and AG-401 produces synergistic inhibition for osteosarcoma in patient-derived xenograft (PDX) models. Our findings demonstrate that NAT10 facilitates osteosarcoma progression through the ATF4/ASNS/Asn axis, and pharmacological inhibition of NAT10 may be a feasible therapeutic approach for osteosarcoma.


Subject(s)
Activating Transcription Factor 4 , Asparagine , Aspartate-Ammonia Ligase , Osteosarcoma , Humans , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/genetics , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Animals , Cell Line, Tumor , Aspartate-Ammonia Ligase/metabolism , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/antagonists & inhibitors , Mice , Asparagine/metabolism , Disease Progression , Xenograft Model Antitumor Assays , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Cell Proliferation/drug effects , Mice, Nude , Male , Female
10.
Cancer Med ; 13(18): e70248, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39315544

ABSTRACT

INTRODUCTION: Osteosarcoma is the most common primary malignancy of the bone. There is a lack of effective treatments for patients who experience relapsed osteosarcoma. One treatment for relapsed patients is gemcitabine and docetaxel combination chemotherapy (GEMDOX). This systematic review aimed to establish the efficacy of this chemotherapy regimen, as well as identify the common severe toxicities that are associated with it. Resistant osteosarcoma cell lines developed from MG-63 and HOS-143B were used to represent relapsed osteosarcoma patients in a pre-clinical study. RESULTS: We identified 11 retrospective and Phase II studies that were suitable for inclusion in our review. 10.65% of patients had a response to gemcitabine and docetaxel combination therapy and the disease control rate was 35% (n = 197). 36%, 35.3% and 18.04% of patients experienced grade 3 or 4 neutropenia, thrombocytopenia and anaemia respectively (n = 133). Male patients (X2 = 9.14, p < 0.05) and those below the age of 18 (X 2 = 10.94, p < 0.05) responded better to GEMDOX treatment than females and patients older than 18 years. The resistant osteosarcoma cell lines remained sensitive to either single-agent gemcitabine, docetaxel, and the combination of both. Cisplatin-resistant models (MG-63/CISR8 & HOS-143B/CISR8) were the most responsive to GEMDOX treatment compared to doxorubicin, methotrexate, and triple-combination resistant models. CONCLUSION: GEMDOX treatment has potential efficacy in relapsed osteosarcoma patients especially those with cisplatin resistance. To directly compare the efficacy of GEMDOX therapy against other therapies randomised phase III clinical trials with adequate patient follow up must be performed to improve treatment options for osteosarcoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Bone Neoplasms , Deoxycytidine , Docetaxel , Drug Resistance, Neoplasm , Gemcitabine , Osteosarcoma , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Humans , Docetaxel/therapeutic use , Docetaxel/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Deoxycytidine/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Taxoids/therapeutic use , Taxoids/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Cell Line, Tumor , Male , Female , Treatment Outcome , Animals
11.
Int J Biol Sci ; 20(11): 4114-4127, 2024.
Article in English | MEDLINE | ID: mdl-39247831

ABSTRACT

Osteosarcoma is the most prevalent form of primary malignant bone tumor, primarily affecting children and adolescents. The nerve growth factors (NGF) referred to as neurotrophins have been associated with cancer-induced bone pain; however, the role of NGF in osteosarcoma has yet to be elucidated. In osteosarcoma samples from the Genomic Data Commons data portal, we detected higher levels of NGF and M2 macrophage markers, but not M1 macrophage markers. In cellular experiments, NGF-stimulated osteosarcoma conditional medium was shown to facilitate macrophage polarization from the M0 to the M2 phenotype. NGF also enhanced VCAM-1-dependent monocyte adhesion within the osteosarcoma microenvironment by down-regulating miR-513c-5p levels through the FAK and c-Src cascades. In in vivo xenograft models, the overexpression of NGF was shown to enhance tumor growth, while the oral administration of the TrK inhibitor larotrectinib markedly antagonized NGF-promoted M2 macrophage expression and tumor progression. These results suggest that larotrectinib could potentially be used as a therapeutic agent aimed at mitigating NGF-mediated osteosarcoma progression.


Subject(s)
Monocytes , Nerve Growth Factor , Osteosarcoma , Tumor Microenvironment , Vascular Cell Adhesion Molecule-1 , Osteosarcoma/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Humans , Nerve Growth Factor/metabolism , Animals , Tumor Microenvironment/drug effects , Monocytes/metabolism , Monocytes/drug effects , Vascular Cell Adhesion Molecule-1/metabolism , Mice , Cell Adhesion/drug effects , Cell Line, Tumor , Bone Neoplasms/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Macrophages/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Mice, Nude
12.
J Nanobiotechnology ; 22(1): 566, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39272206

ABSTRACT

Breast cancer is the most common malignant tumor that threatens women's life and health, and metastasis often occurs in the advanced stage of breast cancer, leading to pathological bone destruction and seriously reducing patient quality of life. In this study, we coupled chlorin e6 (Ce6) with mono-(6-amino-6-deoxy)-beta-cyclodextrin (ß-CD) to form Ce6-CD, and combined ferrocene with the FFVLG3C peptide and PEG chains to form the triblock molecule Fc-pep-PEG. In addition, the IDO-1 inhibitor NLG919 was loaded with Ce6-CD and Fc-pep-PEG to construct the supramolecular nanoparticle NLG919@Ce6-CD/Fc-pep-PEG (NLG919@CF). After laser irradiation, Ce6 produced robust reactive oxidative species to induce tumor cell apoptosis. Simultaneously, ferrocene became charged, and Fc-pep-PEG dissociated from the spherical nanoparticles, enabling their transformation into nanofibers, which increased both the retention effect and the induction of ferroptosis. The released NLG919 reduced the number of regulatory T cells (Tregs) and restored the function of cytotoxic T lymphocytes (CTLs) by inhibiting the activity of IDO-1. Moreover, combined administration with an anti-PD-1 antibody further relieved immune suppression in the tumor microenvironment. This article presents a new strategy for the clinical treatment of breast cancer with bone metastasis and osteolysis.


Subject(s)
Bone Neoplasms , Breast Neoplasms , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Bone Neoplasms/secondary , Bone Neoplasms/drug therapy , Humans , Animals , Mice , Cell Line, Tumor , Porphyrins/chemistry , Porphyrins/therapeutic use , Porphyrins/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Chlorophyllides , Mice, Inbred BALB C , Apoptosis/drug effects , Nanostructures/chemistry , Nanostructures/therapeutic use , Nanoparticles/chemistry , Ferrous Compounds/chemistry , Ferrous Compounds/therapeutic use , Immunosuppression Therapy/methods
13.
Drug Deliv ; 31(1): 2391001, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39239763

ABSTRACT

A common malignant bone neoplasm in teenagers is Osteosarcoma. Chemotherapy, surgical therapy, and radiation therapy together comprise the usual clinical course of treatment for Osteosarcoma. While Osteosarcoma and other bone tumors are typically treated surgically, however, surgical resection frequently fails to completely eradicate tumors, and in turn becomes the primary reason for postoperative recurrence and metastasis, ultimately leading to a high rate of mortality. Patients still require radiation and/or chemotherapy after surgery to stop the spread of the tumor and its metastases, and both treatments have an adverse influence on the body's organ systems. In the postoperative management of osteosarcoma, bone scaffolds can load cargos (growth factors or drugs) and function as drug delivery systems (DDSs). This review describes the different kinds of bone scaffolds that are currently available and highlights key studies that use scaffolds as DDSs for the treatment of osteosarcomas. The discussion also includes difficulties and perspectives regarding the use of scaffold-based DDSs. The study may serve as a source for outlining efficient and secure postoperative osteosarcoma treatment plans.


Subject(s)
Bone Neoplasms , Drug Delivery Systems , Osteosarcoma , Tissue Scaffolds , Osteosarcoma/drug therapy , Humans , Drug Delivery Systems/methods , Bone Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Bone and Bones/drug effects , Animals
14.
ACS Biomater Sci Eng ; 10(9): 5832-5843, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39121344

ABSTRACT

Chondrosarcoma (CHS), also known as malignant cartilage tumors, is the second most common bone cancer after osteosarcoma. This tumor is particularly chemo- and radioresistant, and the only therapeutic alternative is surgery with wide margins. The tumor immune microenvironment reveals an infiltration of tumor-associated macrophages (TAMs) sometimes approaching 50% of the tumor mass, mainly differentiated into M2-like phenotype and correlated with poor prognosis and metastasis. Thus, macrophage-targeting therapies could have an interest in the management of CHS. To evaluate these strategies, we propose here the development of a three-dimensional (3D) tumoroid co-culture model between two human CHS cell lines (JJ012 and CH2879) and a human leukemia monocytic cell line (THP-1) in a methylcellulose matrix. These two models were compared to the in vivo xenograft models in terms of macrophage phenotypes, proteoglycans, MMP-9, and COX-2 expression. Finally, mifamurtide, an immunomodulator acting on TAMs, was evaluated on the most in vitro relevant model: 3D co-culture CH2879 model. Our results showed that it is now possible to develop 3D models that very accurately mimic what is found in vivo with the possibility of evaluating treatments specific to a tumor cell component.


Subject(s)
Chondrosarcoma , Coculture Techniques , Humans , Chondrosarcoma/pathology , Chondrosarcoma/drug therapy , Animals , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Cell Line, Tumor , Mice , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/immunology , Tumor Microenvironment/drug effects , Proteoglycans , Matrix Metalloproteinase 9/metabolism , Antineoplastic Agents/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism
15.
Cancer Med ; 13(15): e70077, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39101490

ABSTRACT

AIMS: Bone cancer produces severe pain that is treated with opioids, but serious side effects limit opioid utilization. There is therefore a need to develop effective and safe non-opioid alternatives. The lipid mediator, Resolvin D1 (RvD1), could be a prospective candidate for cancer pain treatment. To assess RvD1 and other potential candidates, appropriate animal models that recapitulate clinical features must be used. Although several preclinical models of cancer pain have been developed, the influence of sex on the development of cancer pain and the effectiveness of RvD1 have not been studied. RESULTS: Using a mouse model of fibrosarcoma growth in and around the calcaneus bone, we demonstrated that the mechanical hyperalgesia in the tumor-bearing hind paw develops independently of sex, except that it developed a little sooner in female mice. A single intravenous injection of RvD1 (0.001-10 µg/kg) decreased hyperalgesia in both sexes with similar potency (ED50 = 0.0015 µg/kg) and efficacy. Repeated daily administration of 10 µg/kg RvD1 prolonged the analgesic effect and completely abolished hyperalgesia. This was also independent of sex. CONCLUSION: In this preclinical mouse model of bone cancer pain, the development of pain and the analgesic effectiveness of RvD1 are not influenced by sex.


Subject(s)
Bone Neoplasms , Cancer Pain , Disease Models, Animal , Docosahexaenoic Acids , Hyperalgesia , Animals , Female , Bone Neoplasms/drug therapy , Bone Neoplasms/complications , Bone Neoplasms/secondary , Male , Cancer Pain/drug therapy , Cancer Pain/etiology , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/pharmacology , Mice , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Analgesics/pharmacology , Analgesics/administration & dosage , Fibrosarcoma/drug therapy , Fibrosarcoma/pathology , Fibrosarcoma/complications , Sex Factors , Pain Measurement
16.
Cancer Epidemiol ; 92: 102653, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39179493

ABSTRACT

INTRODUCTION: Ewing sarcoma is an aggressive malignancy primarily affecting children and adolescents. Limited research is available on treatment practices, clinical course, and survival in adults. METHODS: A multi-institution retrospective cohort study of all adults (>18 years) and children (≤18 years) with Ewing sarcoma treated in British Columbia, Canada between January 01, 2000 and December 31, 2018. RESULTS: One-hundred seven individuals (66 adults, 41 children) were included in the analysis. 5-year OS was 58 % in adults and 75 % in children. For individuals with local disease, 5-year OS was 74 % in adults and 84 % in children. Adult status was associated with impaired PFS (HR, 1.8; 95 % CI, 1.0 - 3.1, p=0.04) and OS (HR, 1.8; 95 % CI, 0.9 - 3.5; p=0.088). A Charlson Comorbidity Index (CCI) ≥3 was associated with impaired survival in adults and children (HR, 3.9, 95 % CI, 2.0 - 7.5; p=<0.001); baseline CCIs were not significantly different between groups. Most adults (61/66; 92 %) and all children (41/41; 100 %) received systemic treatment with no significant difference in mean lines of therapy, treatment modalities or agents. Most children received interval-compressed chemotherapy (35/41; 85 %) compared to adults (19/61; 29 %; p=<0.001). Interval-compression was not significantly associated with improved survival in adults with local disease (HR, 0.51; 95 % CI 0.1 - 2.3; p=0.373). Children more often initiated treatment within 28 days of diagnosis (31/33; 94 %) compared to adults (41/64; 64 %, p=0.001). Treatment within 28 days was associated with improved survival in the entire cohort (HR, 2.04 95 % CI, 1.1 - 3.9; p = 0.03). This association was preserved in subanalysis of individuals with local disease (HR, 5.4; 95 % CI, 1.9 - 15; p = 0.001) and only adults (HR, 5.3, 95 % CI, 1.7 - 17; p = 0.005). DISCUSSION: Survival for adults with Ewing sarcoma is inferior to children despite similarities in presentation, tumour characteristics and treatments. Further studies on the value of interval-compression in adults are required. Timely initation of treatment should be a priority for this disease.


Subject(s)
Sarcoma, Ewing , Humans , Sarcoma, Ewing/mortality , Sarcoma, Ewing/therapy , Sarcoma, Ewing/pathology , Sarcoma, Ewing/drug therapy , Male , Female , Retrospective Studies , Adult , Adolescent , Child , Young Adult , Middle Aged , British Columbia/epidemiology , Child, Preschool , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/therapy , Survival Rate , Treatment Outcome , Aged
17.
Biomed Pharmacother ; 179: 117284, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39151310

ABSTRACT

Osteosarcoma is the most common primary bone malignancy with a challenging prognosis marked by a high rate of metastasis. The limited success of current treatments may be partially attributed to an incomplete understanding of osteosarcoma pathophysiology and to the absence of reliable in vitro models to select the best molecules for in vivo studies. Among the natural compounds relevant for osteosarcoma treatment, Licochalcone A (Lic-A) and chalcone derivatives are particularly interesting. Here, Lic-A and selected derivatives have been evaluated for their anticancer effect on multicellular tumor spheroids from MG63 and 143B osteosarcoma cell lines. A metabolic activity assay revealed Lic-A, 1i, and 1k derivatives as the most promising candidates. To delve into their mechanism of action, caspase activity assay was conducted in 2D and 3D in vitro models. Notably, apoptosis and autophagic induction was generally observed for Lic-A and 1k. The invasion assay demonstrated that Lic-A and 1k possess the ability to mitigate the spread of osteosarcoma cells within a matrix. The effectiveness of chalcone as a natural scaffold for generating potential antiproliferative agents against osteosarcoma has been demonstrated. In particular, chalcones exert their antiproliferative activity by inducing apoptosis and autophagy, and in addition they are capable of reducing cell invasion. These findings suggest Lic-A and 1k as promising antitumor agents against osteosarcoma cells.


Subject(s)
Apoptosis , Autophagy , Bone Neoplasms , Chalcones , Osteosarcoma , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Humans , Chalcones/pharmacology , Autophagy/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Proliferation/drug effects , Neoplasm Invasiveness , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Antineoplastic Agents/pharmacology
18.
Discov Med ; 36(187): 1648-1656, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39190380

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is commonly recognized as a malignant cancer originating from bone-forming mesenchymal stem cells, comprising approximately 20% of sarcomas. Baicalin, a bioactive flavonoid glycoside isolated from Scutellaria baicalensis, has been demonstrated to possess potent anti-inflammatory and neuroprotective properties. OBJECTIVE: To explore the potential mechanisms through which baicalin exerts anti-osteosarcoma effects and facilitates osteogenesis in vitro. METHODS: Cell Counting Kit-8 (CCK-8), scratch assay, and transwell assay were employed to assess the effects of baicalin at varying concentrations (20, 40, and 80 µM) on U2OS cell proliferation, invasion, and migration, respectively. Western blot and qRT-PCR analyses were conducted to evaluate the influence of baicalin on the osteogenic potential of OS cells by examining osteoblast markers such as osteocalcin (OCN), osteopontin (OPN), and runt-related transcription factor 2 (RUNX2), as well as the osteoclast marker-receptor activator of nuclear factor kappa B ligand (RANKL). Additionally, the impact of baicalin on epithelial-mesenchymal transition (EMT) markers (N-cadherin, E-cadherin, Vimentin) and proteins related to the Nuclear factor κB (NF-κB) signaling pathway (p-p65, p-IκBα, p65, IκBα) in OS cells was evaluated via western blot analysis. The activity and mineralization capacity of Alkaline Phosphatase (ALP) in baicalin-treated cells were examined through ALP staining and Alizarin Red S (ARS) staining. RESULTS: Baicalin exhibited significant suppression of OS cell U2OS invasion (p < 0.01), migration (p < 0.01), and proliferation (p < 0.05) at various concentrations. Additionally, baicalin treatment notably increased the E-cadherin protein level, while decreasing the expression levels of Vimentin and N-cadherin proteins (p < 0.01), thus promoting EMT. Following baicalin treatment, there was a marked elevation in the protein and mRNA expression levels of RUNX2, OPN, and OCN, while the expression level of RANKL protein was reduced (p < 0.05), indicating enhanced osteogenic differentiation. The groups treated with baicalin exhibited higher ALP activity and mineralization ability (p < 0.01). Moreover, baicalin treatment significantly reduced the expression levels of p-IκBα and p-p65 proteins, as well as the ratios of p-IκBα/IκBα and p-p65/p65 (p < 0.01). These effects of baicalin were concentration-dependent, with higher concentrations yielding stronger effects. CONCLUSION: In vitro, baicalin demonstrates anti-OS effects and facilitates osteogenic differentiation, potentially by inhibiting NF-κB pathway activity.


Subject(s)
Cell Differentiation , Cell Proliferation , Epithelial-Mesenchymal Transition , Flavonoids , NF-kappa B , Osteogenesis , Osteosarcoma , Signal Transduction , Humans , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/drug therapy , Flavonoids/pharmacology , Osteogenesis/drug effects , NF-kappa B/metabolism , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Cell Line, Tumor , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/drug therapy , Cell Movement/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics
19.
Cancer Control ; 31: 10732748241274188, 2024.
Article in English | MEDLINE | ID: mdl-39183728

ABSTRACT

BACKGROUND: The standard treatment for localized osteosarcoma is neoadjuvant chemotherapy before surgery, followed by adjuvant chemotherapy. Our aim was to report the rate of histopathological response to neoadjuvant chemotherapy for the treatment of extremity osteosarcoma in Vietnam. METHODS: We performed a retrospective study of stage II conventional osteosarcoma patients under 40 years-old who received MAP regimen as neoadjuvant chemotherapy at the Vietnam National Cancer Hospital between June 2019 and June 2022. Histopathological response was evaluated using the Huvos grading system, in which a good histopathological response was defined as a necrotic rate of 90% or more. RESULTS: Thirty-five eligible patients were included in the study. Male patients accounted for 65.7%, with a median age of 16 years (range, 8-38 years). Of the 35 cases, 31 were reported as stage IIB (88.6%). The femur and tibia were the most common sites in our study, accounting for 51.4% and 34.3%, respectively. The most common pathologic subtype was osteoblastic osteosarcoma (68.6%), followed by chondroblastic subtype (20%). After two cycles of MAP-regimen neoadjuvant chemotherapy, 28 of 35 patients (80%) underwent limb-sparing surgery. A good histopathological response was observed in 18 of 35 patients (51.4%). There were significant correlations between the duration of symptoms (P = 0.016), LDH (P = 0.001) serum levels at initial presentation, and ALP (P = 0.043) serum levels at initial presentation with histopathological response. CONCLUSION: This retrospective study suggests a possible association between symptom duration, pre-treatment LDH levels, and pre-treatment ALP levels with histopathological response rates. Additional clinical investigations with long-term follow-up are needed to investigate survival outcomes in the Asian population.


Subject(s)
Bone Neoplasms , Neoadjuvant Therapy , Osteosarcoma , Humans , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/therapy , Osteosarcoma/mortality , Male , Neoadjuvant Therapy/methods , Retrospective Studies , Adult , Adolescent , Vietnam , Young Adult , Female , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/therapy , Bone Neoplasms/mortality , Child , Chemotherapy, Adjuvant/methods , Chemotherapy, Adjuvant/statistics & numerical data , Neoplasm Staging , Extremities/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
20.
Stomatologiia (Mosk) ; 103(4): 33-36, 2024.
Article in Russian | MEDLINE | ID: mdl-39171341

ABSTRACT

THE AIM OF THE STUDY: Was to investigate the dynamics of mandibular density in cancer patients during therapy with zolendronic acid. MATERIALS AND METHODS: The study comprised 14 patients who received zolendronic acid at a dosage of 4 mg once every 28 days for bone metastases. In all 14 patients, measurements of mandibular density values on CT scans were performed over time. RESULTS: Using multiple linear regression analysis, a model was developed to predict the effect of the number of zolendronic acid injections «X1¼ on the dynamics of mandibular density «Y¼. The resulting formula for predicting mandibular density is Y = 5.9 X1 + 49 HU. CONCLUSION: The model has limitations due to the study design but still can be used by oncologists and dentists to assess mandibular density in patients taking zolendronic acid.


Subject(s)
Bone Density Conservation Agents , Diphosphonates , Imidazoles , Mandible , Zoledronic Acid , Humans , Female , Mandible/drug effects , Mandible/diagnostic imaging , Zoledronic Acid/administration & dosage , Zoledronic Acid/pharmacology , Male , Imidazoles/administration & dosage , Imidazoles/pharmacology , Middle Aged , Diphosphonates/administration & dosage , Diphosphonates/pharmacology , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/therapeutic use , Bone Density Conservation Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Bone Density/drug effects , Aged , Tomography, X-Ray Computed , Adult
SELECTION OF CITATIONS
SEARCH DETAIL