Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.067
Filter
1.
J Cancer Res Ther ; 20(3): 979-983, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-39023606

ABSTRACT

BACKGROUND: There are limited data on the efficacy of targeted therapy in metastatic osteosarcoma. The goal of this study was to assess the effectiveness of sorafenib in adult patients with heavily pretreated metastatic osteosarcoma. METHOD: Patients with metastatic osteosarcoma aged more than 18 years were assessed retrospectively. The patients' clinical, pathological, and therapeutic data were collected. For survival analysis, Kaplan-Meier models were used. RESULTS: The research involved 15 patients. The ratio of male and female patients was 2/1, with a median age of 25 years (range: 19-64 years). The most common primary tumor localization was the extremities (66.6%). Fourteen (93.3%) patients had previously received palliative chemotherapy and six (40%) patients had palliative radiotherapy. The median progression-free survival was found as 5.5 months (95% confidence interval, 1.3-9.7). A stable response was observed in seven (46.6%) patients and progressive disease in eight (53.4%) patients. Grade 1-2 toxicities were detected in 50% of the patients, while grade 3-4 toxicities were observed in 14.3% of the patients. CONCLUSIONS: We demonstrated real-life results of sorafenib for disease management in pretreated adult patients with metastatic osteosarcoma in the study. Sorafenib was effective for disease control and well tolerated in the patients. Sorafenib may be a treatment option for disease control after the disease progresses with chemotherapy in patients with metastatic osteosarcoma.


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Osteosarcoma , Sorafenib , Humans , Osteosarcoma/drug therapy , Osteosarcoma/mortality , Osteosarcoma/pathology , Sorafenib/therapeutic use , Sorafenib/adverse effects , Female , Male , Adult , Young Adult , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Middle Aged , Retrospective Studies , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , Treatment Outcome , Neoplasm Metastasis
2.
J Orthop Surg Res ; 19(1): 405, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39010095

ABSTRACT

BACKGROUND: Currently, there is limited understanding regarding the clinical significance of the tumor-stroma ratio (TSR) in giant cell tumor of bone (GCTB). Hence, we aimed to investigate the distribution of TSR in GCTB and explore its correlation with various clinicopathologic factors, immune microenvironment, survival prognosis, and denosumab treatment responsiveness. METHODS: We conducted a multicenter cohort study comprising 426 GCTB patients treated at four centers. TSR was evaluated on hematoxylin and eosin-stained and immunofluorescent sections of tumor specimens. Immunohistochemistry was performed to assess CD3+, CD4+, CD8+, CD20+, PD-1+, PD-L1+, and FoxP3+ TIL subtypes as well as Ki-67 expression levels in 426 tissue specimens. These parameters were then analyzed for their correlations with patient outcomes [local recurrence-free survival (LRFS) and overall survival (OS)], clinicopathological features, and denosumab treatment responsiveness. RESULTS: Low TSR was significantly associated with poor LRFS and OS in both cohorts. Furthermore, TSR was also correlated with multiple clinicopathological features, TIL subtype expression, and denosumab treatment responsiveness. TSR demonstrated similar predictive capabilities as the conventional Campanacci staging system for predicting patients' LRFS and OS. CONCLUSION: The results of this study provide evidence supporting the use of TSR as a reliable prognostic tool in GCTB and as a predictor of denosumab treatment responsiveness. These findings may aid in developing individualized treatment strategies for GCTB patients in the future.


Subject(s)
Bone Neoplasms , Denosumab , Giant Cell Tumor of Bone , Tumor Microenvironment , Humans , Denosumab/therapeutic use , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/pathology , Tumor Microenvironment/immunology , Female , Male , Adult , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/immunology , Middle Aged , Cohort Studies , Young Adult , Treatment Outcome , Prognosis , Bone Density Conservation Agents/therapeutic use , Adolescent
3.
Front Endocrinol (Lausanne) ; 15: 1415722, 2024.
Article in English | MEDLINE | ID: mdl-39015175

ABSTRACT

Osteosarcoma is a common malignancy that often occurs in children, teenagers and young adults. Although the treatment strategy has improved, the results are still poor for most patients with metastatic or recurrent osteosarcomas. Therefore, it is necessary to identify new and effective prognostic biomarkers and therapeutic targets for diseases. Human genomes contain lncRNAs, transcripts with limited or insufficient capacity to encode proteins. They have been implicated in tumorigenesis, particularly regarding the onset, advancement, resistance to treatment, recurrence and remote dissemination of malignancies. Aberrant lncRNA expression in osteosarcomas has been reported by numerous researchers; lncRNAs have the potential to exhibit either oncogenic or tumor-suppressing behaviors and thus, to govern the advancement of this skeletal cancer. They are suspected to influence osteosarcoma cell growth, replication, invasion, migration, remote dissemination and programmed cell death. Additionally, they have been recognized as clinical markers, and may participate in the development of multidrug resistance. Therefore, the study of lncRNAs in the growth, metastasis, treatment and prognosis of osteosarcoma is very important for the active prevention and treatment of osteosarcoma. Consequently, this work reviews the functions of lncRNAs.


Subject(s)
Biomarkers, Tumor , Bone Neoplasms , Drug Resistance, Neoplasm , Osteosarcoma , RNA, Long Noncoding , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/diagnosis , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Humans , RNA, Long Noncoding/genetics , Drug Resistance, Neoplasm/genetics , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/diagnosis , Gene Expression Regulation, Neoplastic
4.
Oncol Res ; 32(7): 1163-1172, 2024.
Article in English | MEDLINE | ID: mdl-38948019

ABSTRACT

Background: Osteosarcoma is the most common malignant primary bone tumor. The prognosis for patients with disseminated disease remains very poor despite recent advancements in chemotherapy. Moreover, current treatment regimens bear a significant risk of serious side effects. Thus, there is an unmet clinical need for effective therapies with improved safety profiles. Taurolidine is an antibacterial agent that has been shown to induce cell death in different types of cancer cell lines. Methods: In this study, we examined both the antineoplastic and antiangiogenic effects of taurolidine in animal models of osteosarcoma. K7M2 murine osteosarcoma cells were injected, both intramuscular and intraperitoneal, into 60 BALB/c mice on day zero. Animals were then randomized to receive treatment with taurolidine 2% (800 mg/kg), taurolidine 1% (400 mg/kg), or NaCl 0.9% control for seven days by intravenous or intraperitoneal administration. Results: After 35 days, mice were euthanized, and the tumors were harvested for analysis. Eighteen mice were excluded from the analysis due to complications. Body weight was significantly lower in the 2% taurolidine intraperitoneal treatment group from day 9 to 21, consistent with elevated mortality in this group. Intraperitoneal tumor weight was significantly lower in the 1% (p = 0.003) and 2% (p = 0.006) intraperitoneal taurolidine treatment groups compared to the control. No antineoplastic effects were observed on intramuscular tumors or for intravenous administration of taurolidine. There were no significant differences in microvessel density or mitotic rate between treatment groups. Reduced body weight and elevated mortality in the 2% taurolidine intraperitoneal group suggest that the lower 1% dose is preferable. Conclusions: In conclusion, there is no evidence of antiangiogenic activity, and the antitumor effects of taurolidine on osteosarcoma observed in this study are limited. Moreover, its toxic profile grants further evaluation. Given these observations, further research is necessary to refine the use of taurolidine in osteosarcoma treatment.


Subject(s)
Bone Neoplasms , Disease Models, Animal , Osteosarcoma , Taurine , Thiadiazines , Tumor Burden , Animals , Taurine/analogs & derivatives , Taurine/pharmacology , Taurine/therapeutic use , Thiadiazines/pharmacology , Thiadiazines/therapeutic use , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/blood supply , Mice , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Tumor Burden/drug effects , Microvascular Density/drug effects , Mice, Inbred BALB C , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Humans , Neovascularization, Pathologic/drug therapy
5.
Front Immunol ; 15: 1379056, 2024.
Article in English | MEDLINE | ID: mdl-38957472

ABSTRACT

Background: Bone metastases (BoMs) are prevalent in patients with metastatic non-small-cell lung cancer (NSCLC) however, there are limited data detailing how BoMs respond to immune checkpoint inhibitors (ICIs). The purpose of this study was to compare the imaging response to ICIs of BoMs against visceral metastases and to evaluate the effect of BoMs on survival. Materials and methods: A retrospective, multicentre cohort study was conducted in patients with NSCLC treated with nivolumab or pembrolizumab in Alberta, Canada from 2015 to 2020. The primary endpoint was the real-world organ specific progression free survival (osPFS) of bone versus visceral metastases. Visceral metastases were categorized as adrenal, brain, liver, lung, lymph node, or other intra-abdominal lesions. The secondary outcome was overall survival (OS) amongst patients with and without BoMs. Results: A total of 573 patients were included of which all patients had visceral metastases and 243 patients (42.4%) had BoMs. High PD-L1 expression was identified in 268 patients (46.8%). No significant difference in osPFS was observed between bone, liver, and intra-abdominal metastases (p=0.20 and p=0.76, respectively), with all showing shorter osPFS than other disease sites. There was no difference in the osPFS of extra-thoracic sites of disease in patients with high PD-L1 expression. There was significant discordance between visceral disease response and bone disease response to ICI (p=0.047). The presence of BoMs was an independent poor prognostic factor for OS (HR 1.26, 95%CI: 1.05-1.53, p=0.01). Conclusion: Metastatic bone, liver, and intra-abdominal lesions demonstrated inferior clinical responses to ICI relative to other sites of disease. Additionally, the presence of bone and liver metastases were independent poor prognostic factors for overall survival. This real-world data suggests that BoMs respond poorly to ICI and may require treatment adjuncts for disease control.


Subject(s)
Bone Neoplasms , Carcinoma, Non-Small-Cell Lung , Immune Checkpoint Inhibitors , Lung Neoplasms , Humans , Immune Checkpoint Inhibitors/therapeutic use , Male , Female , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Aged , Retrospective Studies , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Lung Neoplasms/mortality , Middle Aged , Bone Neoplasms/secondary , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Aged, 80 and over , Antibodies, Monoclonal, Humanized/therapeutic use , Adult , Treatment Outcome
6.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000064

ABSTRACT

Chondrosarcoma (CS) is a rare malignant bone sarcoma that primarily affects cartilage cells in the femur and pelvis. While most subtypes exhibit slow growth with a very good prognosis, some aggressive subtypes have a poorer overall survival. CS is known for its resistance to chemotherapy and radiotherapy, leaving surgery as the sole effective therapeutic option. Cold physical plasma (CPP) has been explored in vitro as a potential therapy, demonstrating positive anti-tumor effects on CS cells. This study investigated the synergistic effects of combining CPP with cytostatics on CS cells. The chemotherapeutic agents cisplatin, doxorubicin, and vincristine were applied to two CS cell lines (CAL-78 and SW1353). After determining their IC20 and IC50, they were combined with CPP in both cell lines to assess their impact on the cell proliferation, viability, metabolism, and apoptosis. This combined approach significantly reduced the cell proliferation and viability while increasing the apoptosis signals compared to cytostatic therapy alone. The combination of CPP and chemotherapeutic drugs shows promise in targeting chemoresistant CS cells, potentially improving the prognosis for patients in clinical settings.


Subject(s)
Apoptosis , Bone Neoplasms , Cell Proliferation , Cell Survival , Chondrosarcoma , Doxorubicin , Plasma Gases , Chondrosarcoma/drug therapy , Chondrosarcoma/pathology , Humans , Plasma Gases/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Vincristine/pharmacology , Combined Modality Therapy
7.
Medicine (Baltimore) ; 103(28): e38851, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38996101

ABSTRACT

RATIONALE: Patients with bone metastasis-associated cancer pain often experience a complex mix of pain types. Consequently, the use of multimodal combination therapy is essential. While monitoring for common adverse reactions in pain treatment, it is also crucial to be vigilant for the rare but serious serotonin syndrome. PATIENT CONCERNS: A 53-year-old female with metastatic gastric cancer was hospitalized due to severe, uncontrolled thoracic and cervical pain. During the titration of her cancer pain medication, she developed serotonin syndrome. DIAGNOSES: He was diagnosed with refractory cancer pain and serotonin syndrome. INTERVENTIONS: The complete process of cancer pain medication in a patient with gastric cancer and bone metastasis was analyzed, with a primary focus on the selection of analgesic medications, adjustment of opioid dosages, and prevention and treatment of medication-associated adverse reactions. OUTCOMES: The patient's cancer pain was well controlled, with the prompt management of adverse reactions. Furthermore, by adjusting the medication regimen, intolerable adverse reactions were prevented. LESSONS: In clinical settings, personalized analgesic regimens must be developed for patients with cancer pain to enhance patient compliance with medication, prevent the occurrence of severe adverse reactions, and improve the overall quality of life of patients with cancer. Healthcare professionals should pay increased attention to ADRs associated with opioid medications, whereas pharmacists should assist them in promptly identifying ADRs.


Subject(s)
Bone Neoplasms , Cancer Pain , Pain, Intractable , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Middle Aged , Cancer Pain/drug therapy , Cancer Pain/etiology , Female , Bone Neoplasms/secondary , Bone Neoplasms/drug therapy , Pain, Intractable/etiology , Pain, Intractable/drug therapy , Analgesics, Opioid/therapeutic use , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Analgesics/therapeutic use
8.
JCO Clin Cancer Inform ; 8: e2400078, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39008783

ABSTRACT

PURPOSE: Denosumab is used to treat patients with bone metastasis from solid tumors, but sometimes causes severe hypocalcemia, so careful clinical management is important. This study aims to externally validate our previously developed risk prediction model for denosumab-induced hypocalcemia by using data from two facilities with different characteristics in Japan and to develop an updated model with improved performance and generalizability. METHODS: In the external validation, retrospective data of Kameda General Hospital (KGH) and Miyagi Cancer Center (MCC) between June 2013 and June 2022 were used and receiver operating characteristic (ROC)-AUC was mainly evaluated. A scoring-based updated model was developed using the same data set from a hospital-based administrative database as previously employed. Selection of variables related to prediction of hypocalcemia was based on the results of external validation. RESULTS: For the external validation, data from 235 KGH patients and 224 MCC patients were collected. ROC-AUC values in the original model were 0.879 and 0.774, respectively. The updated model consisting of clinical laboratory tests (calcium, albumin, and alkaline phosphatase) afforded similar ROC-AUC values in the two facilities (KGH, 0.837; MCC, 0.856). CONCLUSION: We developed an updated risk prediction model for denosumab-induced hypocalcemia with small interfacility differences. Our results indicate the importance of using data from plural facilities with different characteristics in the external validation of generalized prediction models and may be generally relevant to the clinical application of risk prediction models. Our findings are expected to contribute to improved management of bone metastasis treatment.


Subject(s)
Databases, Factual , Denosumab , Hypocalcemia , Humans , Hypocalcemia/chemically induced , Hypocalcemia/epidemiology , Hypocalcemia/diagnosis , Denosumab/adverse effects , Denosumab/therapeutic use , Female , Male , Aged , Risk Assessment , Retrospective Studies , Middle Aged , Bone Density Conservation Agents/adverse effects , Japan/epidemiology , ROC Curve , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Aged, 80 and over , Risk Factors
9.
JAMA Netw Open ; 7(7): e2419966, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38980676

ABSTRACT

Importance: The presence of bone pain is significantly associated with worse overall survival (OS) in patients with castration-resistant prostate cancer. However, there are few data regarding bone pain and survival outcomes in the context of metastatic, hormone-sensitive prostate cancer (MHSPC). Objective: To compare survival outcomes among patients with MHSPC by presence or absence of baseline bone pain at diagnosis. Design, Setting, and Participants: This post hoc secondary analysis, conducted from September 1 to December 31, 2023, used patient-level data from SWOG-1216, a phase 3, prospective randomized clinical trial that enrolled patients with newly diagnosed MHSPC from 248 academic and community centers across the US from March 1, 2013, to July 15, 2017. All patients in the intention-to-treat population who had available bone pain status were eligible and included in this secondary analysis. Interventions: In the SWOG-1216 trial, patients were randomized (1:1) to receive either androgen deprivation therapy (ADT) with orteronel, 300 mg orally twice daily (experimental group), or ADT with bicalutamide, 50 mg orally daily (control group), until disease progression, unacceptable toxic effects, or patient withdrawal. Main Outcomes and Measures: Overall survival was the primary end point; progression-free survival (PFS) and prostate-specific antigen (PSA) response were secondary end points. Cox proportional hazards regression models were used for both univariable and multivariable analyses adjusting for age, treatment type, Gleason score, disease volume, Zubrod performance status, and PSA level. Results: Of the 1279 male study participants, 301 (23.5%) had baseline bone pain at MHSPC diagnosis and 896 (70.1%) did not. Bone pain status was unavailable in 82 patients (6.4%). The median age of the 1197 patients eligible and included in this secondary analysis was 67.6 years (IQR, 61.8-73.6 years). Compared with patients who did not experience bone pain, those with baseline bone pain were younger (median age, 66.0 [IQR, 60.1-73.4] years vs 68.2 [IQR, 62.4-73.7] years; P = .02) and had a higher incidence of high-volume disease (212 [70.4%] vs 373 [41.6%]; P < .001). After adjustment, bone pain was associated with shorter PFS and OS. At a median follow-up of 4.0 years (IQR, 2.5-5.4 years), patients with bone pain had median PFS of 1.3 years (95% CI, 1.1-1.7 years) vs 3.7 years (95% CI, 3.3-4.2 years) in patients without initial bone pain (adjusted hazard ratio [AHR], 1.46; 95% CI, 1.22-1.74; P < .001) and OS of 3.9 years (95% CI, 3.3-4.8 years) vs not reached (NR) (95% CI, 6.6 years to NR) in patients without initial bone pain (AHR, 1.66; 95% CI, 1.34-2.05; P < .001). Conclusions and Relevance: In this post hoc secondary analysis of the SWOG-1216 randomized clinical trial, patients with baseline bone pain at MHSPC diagnosis had worse survival outcomes than those without bone pain. These data suggest prioritizing these patients for enrollment in clinical trials, may aid patient counseling, and indicate that the inclusion of bone pain in prognostic models of MHSPC may be warranted. Trial Registration: ClinicalTrials.gov Identifier: NCT01809691.


Subject(s)
Androgen Antagonists , Bone Neoplasms , Prostatic Neoplasms , Humans , Male , Aged , Androgen Antagonists/therapeutic use , Middle Aged , Prostatic Neoplasms/mortality , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/complications , Prostatic Neoplasms/pathology , Bone Neoplasms/secondary , Bone Neoplasms/mortality , Bone Neoplasms/complications , Bone Neoplasms/drug therapy , Nitriles/therapeutic use , Prospective Studies , Cancer Pain/drug therapy , Anilides/therapeutic use , Tosyl Compounds/therapeutic use , Tosyl Compounds/adverse effects , Androstenes/therapeutic use , Pain/drug therapy , Pain/etiology
10.
Cancer Med ; 13(13): e7438, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38967496

ABSTRACT

BACKGROUND: The purpose of this study was to evaluate the efficacy and safety of fruquintinib-based therapy as a salvage therapy for patients with advanced or metastatic sarcoma, including soft tissue sarcoma (STS) and bone sarcoma. METHODS: Patients with advanced or metastatic sarcoma were divided into two groups. One group received fruquintinib monotherapy, while the other received fruquintinib combined therapy. Safety and efficacy of fruquintinib-based therapy were recorded and reviewed retrospectively, including progression-free survival (PFS), overall response rate (ORR), and adverse events (AEs). RESULTS: Between August 2021 and December 2022, 38 sarcoma patients were retrospectively included. A total of 14 patients received fruquintinib alone (including 6 STS and 8 bone sarcoma), while 24 were treated with fruquintinib combined therapy (including 2 STS and 22 bone sarcoma). The median follow-up was 10.2 months (95% CI, 6.4-11.5). For the entire population, the median PFS was 8.0 months (95% CI, 5.5-13.0). The ORR was 13.1%, while the disease control rate (DCR) was 86.8%. The univariate analysis showed that radiotherapy history (HR, 4.56; 95% CI, 1.70-12.24; p = 0.003), bone sarcoma (HR, 0.34; 95% CI, 0.14-0.87; p = 0.024), and treatment method of fruquintinib (HR, 0.36; 95% CI, 0.15-0.85; p = 0.021) were significantly associated with PFS. The multivariate analysis showed that patients without radiotherapy history were associated with a better PFS (HR, 3.71; 95% CI: 1.31-10.55; p = 0.014) than patients with radiotherapy history. Patients in combination group reported pneumothorax (8.3%), leukopenia (33.3%), thrombocytopenia (12.5%), diarrhea (4.2%), and anemia (4.2%) as the most frequent grade 3 or higher treatment-emergent AEs (TEAEs), while there was no severe TEAEs occurred in the monotherapy group. CONCLUSIONS: Fruquintinib-based therapy displayed an optimal tumor control and an acceptable safety profile in patients with advanced or metastatic sarcoma.


Subject(s)
Benzofurans , Bone Neoplasms , Quinazolines , Sarcoma , Humans , Female , Sarcoma/drug therapy , Sarcoma/mortality , Sarcoma/pathology , Male , Middle Aged , Adult , Retrospective Studies , Quinazolines/therapeutic use , Quinazolines/adverse effects , Aged , Benzofurans/therapeutic use , Benzofurans/adverse effects , Bone Neoplasms/secondary , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Young Adult , Salvage Therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Progression-Free Survival , Adolescent , Treatment Outcome
11.
Int J Med Mushrooms ; 26(8): 41-57, 2024.
Article in English | MEDLINE | ID: mdl-38967210

ABSTRACT

Bone metastasis in metastatic breast cancer commonly results in osteolytic lesions due to osteoclast activity, promoting bone destruction and tumor progression. The bioactive fungal isolates, 4-acetyl-antroquinonol B (4-AAQB) and erinacine A, have diverse pharmacological and biological activities. However, their effects on breast cancer bone metastasis treatment remain unclear. Our study aimed to examine the impact of 4-AAQB or erinacine A on breast cancer metastases in bone. The effects of 4-AAQB and erinacine A on breast cancer-induced osteoclastogenesis, breast cancer migration, production of prometastatic cytokine (TGF-ß) and marker (MMP-9), as well as potential MAPK signaling transductions were assessed. The results revealed that 4-AAQB and erinacine A effectively suppressed breast cancer-induced osteoclastogenesis and migration, and reduced TGF-ß and MMP-9 production via Erk or JNK signaling transductions, specifically in breast cancer cells or in breast cancer cells-induced osteoclasts. Based on these findings, either 4-AAQB or erinacine A showed promise in preventing breast cancer metastases in bone.


Subject(s)
Breast Neoplasms , Matrix Metalloproteinase 9 , Osteoclasts , Osteogenesis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Humans , Female , Osteoclasts/drug effects , Osteogenesis/drug effects , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Cell Line, Tumor , Cell Movement/drug effects , Animals , Transforming Growth Factor beta/metabolism , Bone Neoplasms/secondary , Bone Neoplasms/drug therapy , Mice , MAP Kinase Signaling System/drug effects , Cyclohexanones , 4-Butyrolactone/analogs & derivatives
12.
Sci Rep ; 14(1): 16475, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014082

ABSTRACT

Osteosarcoma (OS) is a heterogeneous malignant spindle cell tumor that is aggressive and has a poor prognosis. Although combining surgery and chemotherapy has significantly improved patient outcomes, the prognosis for OS patients with metastatic or recurrent OS has remained unsatisfactory. Therefore, it is imperative to gain a fresh perspective on OS development mechanisms and treatment strategies. After studying single-cell RNA sequencing (scRNA-seq) data in public databases, we identified seven OS subclonal types based on intra-tumor heterogeneity. Subsequently, we constructed a prognostic model based on pro-protein synthesis osteosarcoma (PPS-OS)-associated genes. Correlation analysis showed that the prognostic model performs extremely well in predicting OS patient prognosis. We also demonstrated that the independent risk factors for the prognosis of OS patients were tumor primary site, metastatic status, and risk score. Based on these factors, nomograms were constructed for predicting the 3- and 5-year survival rates. Afterward, the investigation of the tumor immune microenvironment (TIME) revealed the vital roles of γδ T-cell and B-cell activation. Drug sensitivity analysis and immune checkpoint analysis identified drugs that have potential application value in OS. Finally, the jumping translocation breakpoint (JTB) gene was selected for experimental validation. JTB silencing suppressed the proliferation, migration, and invasion of OS cells. Therefore, our research suggests that PPS-OS-related genes facilitate the malignant progression of OS and may be employed as prognostic indicators and therapeutic targets in OS.


Subject(s)
Bone Neoplasms , Osteosarcoma , Tumor Microenvironment , Humans , Osteosarcoma/genetics , Osteosarcoma/therapy , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/mortality , Osteosarcoma/drug therapy , Prognosis , Bone Neoplasms/genetics , Bone Neoplasms/therapy , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/mortality , Bone Neoplasms/drug therapy , Female , Male , Gene Expression Regulation, Neoplastic , Nomograms , Cell Line, Tumor , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation
13.
ACS Appl Mater Interfaces ; 16(27): 34669-34683, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38946103

ABSTRACT

In this research, a novel MgSiO3 fiber membrane (MSFM) loaded with indocyanine green (ICG) and doxorubicin (DOX) was prepared. Because of MgSiO3's unique lamellar structure composed of a silicon-oxygen tetrahedron, magnesium ion (Mg2+) moves easily and can be further replaced with other cations. Therefore, because of the positively charged functional group of ICG, MSFM has a rather high drug loading for ICG. In addition, there is electrostatic attraction between DOX (a cationic drug) and ICG (an anionic drug). Hence, after loading ICG, more DOX can be adsorbed into MSFM because of electrostatic interaction. The ICG endows the MSFM outstanding photothermal therapy (PTT) performance, and DOX as a chemotherapeutic drug can restrain tumor growth. On the one hand, H+ exchanged with the positively charged DOX based on the MgSiO3 special lamellar structure. On the other hand, the thermal effect could break the electrostatic interaction between ICG and DOX. Based on the above two points, both tumor acidic microenvironment and photothermal effect can trigger DOX release. What's more, in vitro and in vivo antiosteosarcoma therapy evaluations displayed a superior synergetic PTT-chemotherapy anticancer treatment and excellent biocompatibility of DOX&ICG-MSFM. Finally, the MSFM was proven to greatly promote cell proliferation, differentiation, and bone regeneration performance in vitro and in vivo. Therefore, MSFM provides a creative perspective in the design of multifunctional scaffolds and shows promising applications in controlled drug delivery, antitumor performance, and osteogenesis.


Subject(s)
Bone Regeneration , Doxorubicin , Indocyanine Green , Osteosarcoma , Doxorubicin/chemistry , Doxorubicin/pharmacology , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Bone Regeneration/drug effects , Animals , Humans , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Mice , Magnesium Silicates/chemistry , Photothermal Therapy , Cell Line, Tumor , Drug Delivery Systems , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Drug Liberation
14.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(4): 541-552, 2024 Apr 28.
Article in English, Chinese | MEDLINE | ID: mdl-39019783

ABSTRACT

OBJECTIVES: Super-enhancer-associated genes may be closely related to the progression of osteosarcoma, curcumin exhibits a certain inhibitory effect on tumors such as osteosarcoma. This study aims to investigate the effects of curcumin on osteosarcoma in vitro and in vivo, and to determine whether curcumin can inhibit the progression of osteosarcoma by suppressing the expression of super-enhancer-associated genes LIM and senescent cell antigen-like-containing domain 1 (LIMS1), secreted protein acidic and rich in cysteine (SPARC), and sterile alpha motif domain containing 4A (SAMD4A). METHODS: Human osteosarcoma cell lines (MG63 cells or U2OS cells) were treated with 5 to 50 µmol/L curcumin for 24, 48, and 72 hours, followed by the methyl thiazolyl tetrazolium (MTT) assay to detect cell viability. Cells were incubated with dimethyl sulfoxide (DMSO) or curcumin (2.5, 5.0 µmol/L) for 7 days, and a colony formation assay was used to measure in vitro cell proliferation. After treatment with DMSO or curcumin (10, 15 µmol/L), a scratch healing assay and a transwell migration assay were performed to evaluate cell migration ability. Real-time reverse transcription polymerase chain reaction (real-time RT-PCR) and Western blotting were used to detect mRNA and protein expression levels of LIMS1, SPARC, and SAMD4A in the cells. An osteosarcoma-bearing nude mouse model was established, and curcumin was administered via gavage for 14 days to assess the impact of curcumin on tumor volume and weight in vivo. Real-time RT-PCR was used to measure mRNA expression levels of LIMS1, SPARC, and SAMD4A in the cancer and adjacent tissues from 12 osteosarcoma patients. RESULTS: After treating cells with different concentrations of curcumin for 24, 48, and 72 hours, cell viability were all significantly decreased. Compared with the DMSO group, the colony formation rates in the 2.5 µmol/L and 5.0 µmol/L curcumin groups significantly declined (both P<0.01). The scratch healing assay showed that, compared with the DMSO group, the migration rates of cells in the 10 µmol/L and 15 µmol/L curcumin groups were significantly reduced. The exception was the 10 µmol/L curcumin group at 24 h, where the migration rate of U2OS cells did not show a statistically significant difference (P>0.05), while all other differences were statistically significant (P<0.01 or P<0.001). The transwell migration assay results showed that the number of migrating cells in the 10 µmol/L and 15 µmol/L curcumin groups was significantly lower than that in the DMSO group (both P<0.001). In the in vivo tumor-bearing mouse experiment, the curcumin group showed a reduction in tumor mass (P<0.01) and a significant reduction in tumor volume (P<0.001) compared with the control group. Compared with the DMSO group, the mRNA expression levels of LIMS1, SPARC, and SAMD4A in the 10 µmol/L and 15 µmol/L curcumin groups were significantly down-regulated (all P<0.05). Additionally, the protein expression level of LIMS1 in U2OS cells in the 10 µmol/L curcumin group was significantly lower than that in the DMSO group (P<0.05). Compared with adjacent tissues, the mRNA expression level of SPARC in osteosarcoma tissues was significantly increased (P<0.001), while the mRNA expression levels of LIMS1 and SAMD4A did not show statistically significant differences (both P>0.05). CONCLUSIONS: Curcumin inhibits the proliferation and migration of osteosarcoma both in vitro and in vivo, which may be associated with the inactivation of super-enhancer-associated gene LIMS1.


Subject(s)
Bone Neoplasms , Cell Movement , Cell Proliferation , Curcumin , Mice, Nude , Osteonectin , Osteosarcoma , Osteosarcoma/genetics , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/metabolism , Curcumin/pharmacology , Humans , Cell Proliferation/drug effects , Cell Movement/drug effects , Animals , Bone Neoplasms/genetics , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Cell Line, Tumor , Mice , Osteonectin/genetics , Osteonectin/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Antineoplastic Agents/pharmacology , Mice, Inbred BALB C
15.
Clin J Oncol Nurs ; 28(3): 247-251, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38830248

ABSTRACT

Androgen deprivation therapy (ADT) is standard, first-line therapy for many aspects of prostate cancer treatment. Although ADT can be an effective treatment to inhibit androgen-fueled cell growth in prostate cancer, suppressi.


Subject(s)
Androgen Antagonists , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/therapy , Androgen Antagonists/therapeutic use , Bone Neoplasms/drug therapy , Advanced Practice Nursing , Aged
16.
Sci Rep ; 14(1): 13451, 2024 06 11.
Article in English | MEDLINE | ID: mdl-38862617

ABSTRACT

Bone-modifying agents (BMA) are extensively used in treating patients with prostate cancer with bone metastases. However, this increases the risk of medication-related osteonecrosis of the jaw (MRONJ). The safety of long-term BMA administration in clinical practice remains unclear. We aimed to determine the cumulative incidence and risk factors of MRONJ. One hundred and seventy-nine patients with prostate cancer with bone metastases treated with BMA at our institution since 2008 were included in this study. Twenty-seven patients (15%) had MRONJ during the follow-up period (median, 19 months; interquartile range, 9-43 months). The 2-year, 5-year, and 10-year cumulative MRONJ incidence rates were 18%, 27%, and 61%, respectively. Multivariate analysis identified denosumab use as a risk factor for MRONJ, compared with zoledronic acid use (HR 4.64, 95% CI 1.93-11.1). Additionally, BMA use at longer than one-month intervals was associated with a lower risk of MRONJ (HR 0.08, 95% CI 0.01-0.64). Furthermore, six or more bone metastases (HR 3.65, 95% CI 1.13-11.7) and diabetes mellitus (HR 5.07, 95% CI 1.68-15.2) were risk factors for stage 2 or more severe MRONJ. MRONJ should be considered during long-term BMA administration in prostate cancer patients with bone metastases.


Subject(s)
Bisphosphonate-Associated Osteonecrosis of the Jaw , Bone Density Conservation Agents , Bone Neoplasms , Denosumab , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Risk Factors , Aged , Incidence , Bisphosphonate-Associated Osteonecrosis of the Jaw/epidemiology , Bisphosphonate-Associated Osteonecrosis of the Jaw/etiology , Denosumab/adverse effects , Bone Neoplasms/secondary , Bone Neoplasms/drug therapy , Bone Neoplasms/epidemiology , Bone Density Conservation Agents/adverse effects , Middle Aged , Zoledronic Acid/adverse effects , Zoledronic Acid/therapeutic use , Aged, 80 and over , Retrospective Studies
17.
Lipids Health Dis ; 23(1): 204, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943207

ABSTRACT

Malignant bone tumors, including primary bone cancer and metastatic bone tumors, are a significant clinical challenge due to their high frequency of presentation, poor prognosis and lack of effective treatments and therapies. Bone tumors are often accompanied by skeletal complications such as bone destruction and cancer-induced bone pain. However, the mechanisms involved in bone cancer progression, bone metastasis and skeletal complications remain unclear. Lysophosphatidic acid (LPA), an intercellular lipid signaling molecule that exerts a wide range of biological effects mainly through specifically binding to LPA receptors (LPARs), has been found to be present at high levels in the ascites of bone tumor patients. Numerous studies have suggested that LPA plays a role in primary malignant bone tumors, bone metastasis, and skeletal complications. In this review, we summarize the role of LPA signaling in primary bone cancer, bone metastasis and skeletal complications. Modulating LPA signaling may represent a novel avenue for future therapeutic treatments for bone cancer, potentially improving patient prognosis and quality of life.


Subject(s)
Bone Neoplasms , Lysophospholipids , Receptors, Lysophosphatidic Acid , Signal Transduction , Humans , Lysophospholipids/metabolism , Bone Neoplasms/secondary , Bone Neoplasms/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Signal Transduction/drug effects , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/genetics , Animals
18.
Biomed Mater ; 19(5)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38917820

ABSTRACT

Metastatic bone lesions are often osteolytic, which causes advanced-stage cancer sufferers to experience severe pain and an increased risk of developing a pathological fracture. Gallium (Ga) ion possesses antineoplastic and anti-bone resorption properties, suggesting the potential for its local administration to impede the growth of metastatic bone lesions. This study investigated the chemotherapeutic potential, cytotoxicity, and osteogenic effects of a Ga-doped glass polyalkenoate cement (GPC) (C-TA2) compared to its non-gallium (C-TA0) counterpart. Ion release profiles revealed a biphasic pattern characterized by an initial burst followed by a gradually declining release of ions. C-TA2 continued to release Ga steadily throughout the experimentation period (7 d) and exhibited prolonged zinc (Zn) release compared to C-TA0. Interestingly, the Zn release from both GPCs appeared to cause a chemotherapeutic effect against H1092 lung cancer cellsin vitro, with the prolonged Zn release from C-TA2 extending this effect. Unfortunately, both GPCs enhanced the viability of HCC2218 breast cancer cells, suggesting that the chemotherapeutic effects of Zn could be tied to cellular differences in preferred Zn concentrations. The utilization of SAOS-2 and MC3T3 cell lines as bone cell models yielded conflicting results, with the substantial decline in MC3T3 viability closely associated with silicon (Si) release, indicating cellular variations in Si toxicity. Despite this ambiguity, both GPCs exhibited harmful effects on the osteogenesis of primary rat osteoblasts, raising concerns about excessive burst Zn release. While Ga/Zn-doped GPCs hold promise for treating metastatic bone lesions caused by lung cancers, further optimization is required to mitigate cytotoxicity on healthy bone.


Subject(s)
Cell Survival , Gallium , Osteogenesis , Gallium/chemistry , Animals , Humans , Cell Line, Tumor , Osteogenesis/drug effects , Cell Survival/drug effects , Mice , Zinc/chemistry , Rats , Glass Ionomer Cements/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Materials Testing , Bone Neoplasms/drug therapy , Osteoblasts/drug effects , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology
19.
Cell Biochem Funct ; 42(4): e4080, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38924104

ABSTRACT

Currently, challenges such as chemotherapy resistance, resulting from preoperative and postoperative chemotherapy, postoperative recurrence, and poor bone regeneration quality, are becoming increasingly prominent in osteosarcoma (OS) treatment. There is an urgent need to find more effective ways to address these issues. Ferroptosis is a novel form of iron-dependent programmed cell death, distinct from other forms of cell death. In this paper, we summarize how, through the three major defense systems of ferroptosis, not only can substances from traditional Chinese medicine, antitumor drugs, and nano-drug carriers induce ferroptosis in OS cells, but they can also be combined with immunotherapy, differentiation therapy, and other treatment modalities to significantly enhance chemotherapy sensitivity and inhibit tumor growth. Thus, ferroptosis holds great potential in treating OS, offering more choices and possibilities for future clinical interventions.


Subject(s)
Bone Neoplasms , Ferroptosis , Osteosarcoma , Ferroptosis/drug effects , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/metabolism , Humans , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Immunotherapy , Animals
20.
Support Care Cancer ; 32(7): 415, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847977

ABSTRACT

PURPOSE: Anemia is relatively common in cancer patients, and is associated with poor survival in patients with various malignancies. However, how anemia would affect prognosis and response to neoadjuvant chemotherapy (NAC) in osteosarcoma (OS) is still without substantial evidence. METHODS: We retrospectively analysed 242 patients with stage II OS around the knee joint in our institute. Changed hemoglobin (Hb) levels (before and after NAC) were recorded to assess the prognostic value in DFS (disease-free survival) and tumor response to NAC. Univariate and multivariate analyses were conducted to identify prognostic factors related with outcome in OS patients. RESULTS: The mean Hb level significantly decreased after NAC (134.5 ± 15.3 g/L vs. 117.4 ± 16.3 g/L). The percentage of mild (21%), moderate (4.2%) and severe (0%) anemia patients markedly increased after NAC: 41%, 24% and 4.1% respectively. There was higher percentage of ≥ 5% Hb decline in patients with tumor necrosis rate < 90% (141 out of 161), compared with those with tumor necrosis rate ≥ 90% (59 out of 81). Further univariate and survival analysis demonstrated that Hb decline had a significant role in prediction survival in OS patients. Patients with ≥ 5% Hb decline after NAC had an inferior DFS compared with those with < 5% Hb decline. CONCLUSION: In osteosarcoma, patients with greater Hb decrease during neoadjuvant treatment were shown to have worse DFS and a poorer response to NAC than those without. Attempts to correct anemia and their effects on outcomes for osteosarcoma patients should be explored in future studies.


Subject(s)
Anemia , Bone Neoplasms , Hemoglobins , Knee Joint , Neoadjuvant Therapy , Osteosarcoma , Humans , Osteosarcoma/drug therapy , Osteosarcoma/mortality , Retrospective Studies , Male , Female , Neoadjuvant Therapy/methods , Hemoglobins/analysis , Adult , Prognosis , Anemia/etiology , Adolescent , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Young Adult , Child , Knee Joint/pathology , Disease-Free Survival , Middle Aged , Multivariate Analysis , Chemotherapy, Adjuvant/methods , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL