Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Adv Mater ; 35(42): e2303718, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37625141

ABSTRACT

Developing a drug delivery platform that possesses universal drug loading capacity to meet various requirements of cancer treatment is a challenging yet interesting task. Herein, a self-assembled gelatin/silk fibroin composite (GSC) particle based drug delivery system is developed via microphase separation followed by desolvation process. Thanks to its preassembled microphase stage, this GSC system is suitable for varying types of drugs. The desolvation process fix drugs inside GSC rapidly and densify the GSC structure, thereby achieving efficient drug loading and providing comprehensive protection for loaded drugs. Actually, the size of this brand-new non-pore dependent drug delivery system can be easily adjusted from 100 nm to 20 µm to fit different scenarios. This work selects GSC with 3 µm diameter as the universal inhaled drug delivery platform, which shows an excellent transmucosal penetration and lung retention ability. Additionally, the MMP-9 sensitive degradation property of GSC enhances the targeted efficiency of drugs and reduces side effects. Intestinally, GSC can self-amplify the regulation of innate immunity to reverse the cancerous microenvironment into an antitumor niche, significantly improving the therapeutic effect of drugs. This study of GSC universal drug platform provides a new direction to develop the next-generation of drug delivery system for lung cancer.


Subject(s)
Fibroins , Lung Neoplasms , Humans , Fibroins/chemistry , Gelatin/chemistry , Matrix Metalloproteinase 9 , Lung Neoplasms/drug therapy , Drug Delivery Systems , Tumor Microenvironment
2.
Nat Commun ; 13(1): 7207, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36418292

ABSTRACT

Osteosarcoma (OS) is a primary malignant bone tumor that most commonly affects children, adolescents, and young adults. Here, we comprehensively analyze genomic, epigenomic and transcriptomic data from 121 OS patients. Somatic mutations are diverse within the cohort, and only TP53 is significantly mutated. Through unsupervised integrative clustering of the multi-omics data, we classify OS into four subtypes with distinct molecular features and clinical prognosis: (1) Immune activated (S-IA), (2) Immune suppressed (S-IS), (3) Homologous recombination deficiency dominant (S-HRD), and (4) MYC driven (S-MD). MYC amplification with HR proficiency tumors is identified with a high oxidative phosphorylation signature resulting in resistance to neoadjuvant chemotherapy. Potential therapeutic targets are identified for each subtype, including platinum-based chemotherapy, immune checkpoint inhibitors, anti-VEGFR, anti-MYC and PARPi-based synthetic lethal strategies. Our comprehensive integrated characterization provides a valuable resource that deepens our understanding of the disease, and may guide future clinical strategies for the precision treatment of OS.


Subject(s)
Bone Neoplasms , Osteosarcoma , Young Adult , Adolescent , Child , Humans , Osteosarcoma/genetics , Osteosarcoma/therapy , Genomics/methods , Transcriptome , Platinum , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics
3.
Orthop Surg ; 14(6): 1161-1166, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35538733

ABSTRACT

OBJECTIVE: To retrospectively analyze and compare the relationship between the success rate of patient-derived xenograft (PDX) modeling of osteosarcoma and prognosis (3-year overall survival rate and disease-free survival rate) and incidence of lung metastasis. METHODS: The sample group consisted of 57 osteosarcoma patients with definite pathological diagnoses from Shanghai General Hospital from 2015-2017. PDX models in 57 patients were analyzed by retrospective analyses. Among the patients currently inoculated, 20 were tumorigenic in the PDX model, and 37 were nontumorigenic. According to the tumorigenicity of PDXs, the corresponding osteosarcoma patients were divided into two groups. The effects of clinically related indicators on the model were retrospectively compared. The patients were followed, and the 3-year survival, 3-year disease-free survival (DFS), and lung metastasis rates were collected. The relationship between the modeling success and patient prognosis was investigated. RESULTS: In the chemotherapy-treated group, the PDX modeling success rate was 17.4%, and in the nonchemotherapy group, the success rate was 47.1%. The success of PDX modeling was related to whether patients received chemotherapy. The success rate of PDX modeling is significantly reduced after receiving chemotherapy. The 3-year overall survival rate of the PDX-grafted group was 49.23%, and that of the PDX-nongrafted group was 65.71%. There was a significant difference between the two groups, showing a strong negative correlation between the 3-year survival rate and the success rate of the PDX model. The 3-year disease-free survival rate of the PDX-grafted group was 29.54%. The 3-year DFS of the PDX-nongrafted group was 50.34%. There was a significant difference between the two groups. Lower grafted rates indicate a higher DFS rate. The incidence of lung metastasis in the PDX-grafted group was 32.4%, and that in the nongrafted group was 13.1%. There was a significant difference between the two groups. The successful establishment of the PDX model indicates that patients are more likely to have lung metastases. CONCLUSIONS: The success of PDX modeling often indicates poor prognosis (low 3-year overall survival rate and disease-free survival rate) and a greater possibility of lung metastasis. Therefore, PDX modeling in osteosarcoma patients can accurately predict the prognosis of patients and the risk of lung metastasis in advance to help us develop better therapeutic strategies.


Subject(s)
Bone Neoplasms , Lung Neoplasms , Osteosarcoma , Animals , Bone Neoplasms/drug therapy , China , Disease Models, Animal , Heterografts , Humans , Osteosarcoma/therapy , Prognosis , Retrospective Studies
4.
Macromol Rapid Commun ; 43(14): e2100926, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35445490

ABSTRACT

Various models of synthetic cells have been developed as researchers have sought to explore the origins of life. Based on the fact that structural complexity is the foundation of higher-order functions, this review focuses on hierarchical structures in synthetic cell models that are inspired by living systems, in which macromolecules are the dominant participants. The underlying advantages and functions provided by biomimetic higher-order structures are discussed from four perspectives, including hierarchical structures in membranes, in the composite construction of membrane-coated artificial cytoplasm, in organelle-like subcellular compartments, as well as in synthetic cell-cell assembled synthetic tissues. In parallel, various feasible driving forces and approaches for the fabrication of such higher-order structures are showcased. Furthermore, both the implemented and potential applications of biomimetic systems, bottom-up biosynthesis, biomedical tissue engineering, and disease therapy are highlighted. This thriving field is gradually narrowing the gap between fundamental research and applied science.


Subject(s)
Artificial Cells , Artificial Cells/chemistry , Biomimetics , Humans , Macromolecular Substances , Tissue Engineering
5.
Front Oncol ; 11: 642134, 2021.
Article in English | MEDLINE | ID: mdl-34168981

ABSTRACT

Osteosarcoma is the most common primary malignant bone tumor, and there are few ideal clinically available drugs. The bromodomain and extraterminal domain (BET) protein is an emerging target for aggressive cancer, but therapies targeting the BET in osteosarcoma have been unsuccessful in clinical trials to date, and further exploration of specific BET inhibitors is of great significance. In our study, we demonstrated that NHWD-870, a potent BET inhibitor in a phase I clinical trial, significantly inhibited tumor proliferation and promoted cell apoptosis by reversing the oncogenic signature in osteosarcoma. More importantly, we identified NHWD-870 impeded binding of BRD4 to the promoter of GP130 leading to diminished activation of JAK/STAT3 signaling pathway. Furthermore, GP130 knockdown significantly sensitizes the chemosensitivity in vitro. In OS cell-derived xenografts, NHWD-870 effectively inhibited the growth of osteosarcoma. Beyond that, NHWD-870 effectively inhibited the differentiation and maturation of precursor osteoclasts in vitro and attenuated osteoclast-mediated bone loss in vivo. Finally, we confirmed the efficacy of synthetic lethal effects of NHWD-870 and cisplatin in antagonizing osteosarcoma in a preclinical PDX model. Taken together, these findings demonstrate that NHWD-870, as an effective BET inhibitor, may be a potential candidate for osteosarcoma intervention linked to its STAT3 signaling inhibitory activity. In addition, NHWD-870 appears to be a promising therapeutic strategy for bone-associated tumors, as it interferes with the vicious cycle of tumor progression and bone destruction.

6.
J Biochem ; 170(2): 275-287, 2021 Oct 11.
Article in English | MEDLINE | ID: mdl-33792698

ABSTRACT

Given the huge cost, long research and development (R&D) time and uncertain side effects of discovering new drugs, drug repositioning of those approved to treat diseases clinically as new drugs for other pathological conditions, especially cancers, is a potential alternative strategy. Disulfiram (DSF), an old drug used to treat alcoholism, has been found to exhibit anticancer activity and improve chemotherapeutic efficacy in cancers by an increasing number of studies. In addition, the combination of DSF and copper may be a more effective therapeutic strategy. In this study, we report the toxicity of the disulfiram/copper (DSF/Cu) complex to human osteosarcoma (OS) both in vitro and in vivo. DSF/Cu significantly inhibited the proliferation and clonogenicity of OS cell lines. Furthermore, the generation of reactive oxygen species (ROS) was triggered by DSF/Cu, and cell arrest, autophagy and apoptosis were induced in an ROS-dependent manner. The underlying mechanism of this process was explored, and DSF/Cu may mainly inhibit OS by inducing apoptosis by activating the ROS/JNK pathway. DSF/Cu also inhibited OS growth in a xenograft model with low levels of organ-related toxicities. These results suggest that the DSF/Cu complex could be an efficient and safe option for the treatment of OS in the clinic.


Subject(s)
Apoptosis/drug effects , Copper/pharmacology , Disulfiram/pharmacology , MAP Kinase Signaling System/drug effects , Osteosarcoma/drug therapy , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Osteosarcoma/metabolism , Osteosarcoma/pathology , Xenograft Model Antitumor Assays
7.
Front Pharmacol ; 12: 798837, 2021.
Article in English | MEDLINE | ID: mdl-35111065

ABSTRACT

Overexpression of the multidrug resistance (MDR)-related protein P-glycoprotein (PGP1), which actively extrudes chemotherapeutic agents from cells and significantly decreases the efficacy of chemotherapy, is viewed as a major obstacle in osteosarcoma chemotherapy. Anlotinib, a novel tyrosine kinase inhibitor (TKI), has good anti-tumor effects in a variety of solid tumors. However, there are few studies on the mechanism of anlotinib reversing chemotherapy resistance in osteosarcoma. In this study, cellular assays were performed in vitro and in vivo to evaluate the MDR reversal effects of anlotinib on multidrug-resistant osteosarcoma cell lines. Drug efflux and intracellular drug accumulation were measured by flow cytometry. The vanadate-sensitive ATPase activity of PGP1 was measured in the presence of a range of anlotinib concentrations. The protein expression level of ABCB1 was detected by Western blotting and immunofluorescence analysis. Our results showed that anlotinib significantly increased the sensitivity of KHOSR2 and U2OSR2 cells (which overexpress PGP1) to chemotherapeutic agents in vitro and in a KHOSR2 xenograft nude mouse model in vivo. Mechanistically, anlotinib increases the intracellular accumulation of PGP1 substrates by inhibiting the efflux function of PGP1 in multidrug-resistant cell lines. Furthermore, anlotinib stimulated the ATPase activity of PGP1 but affected neither the protein expression level nor the localization of PGP1. In animal studies, anlotinib in combination with doxorubicin (DOX) significantly decreased the tumor growth rate and the tumor size in the KHOSR2 xenograft nude mouse model. Overall, our findings suggest that anlotinib may be useful for circumventing MDR to other conventional antineoplastic drugs.

8.
Am J Transl Res ; 12(9): 5882-5907, 2020.
Article in English | MEDLINE | ID: mdl-33042467

ABSTRACT

Osteoporosis is a common metabolic bone disease characterized by low bone mineral density (BMD) and microarchitectural deterioration of bone tissue, which leads to decreased bone strength and increased fracture risk. Osteoporosis mainly results from a disruption of the balance between bone formation mediated by osteoblasts and bone resorption mediated by osteoclasts. At present, the molecular mechanisms underlying osteoporosis are still not fully understood. Long noncoding RNAs (lncRNAs) are RNA molecules that exceed 200 nucleotides (nt) in length and have limited or no protein-coding capacity. Over the past decade, numerous lncRNAs have been demonstrated to participate in multiple biological processes and to play essential roles in the pathogenesis of various diseases. In this review, we summarize recent progress in research on lncRNAs in osteoporosis and mainly focus on their regulatory roles in osteogenesis and osteoclastogenesis. Moreover, we briefly discuss the potential clinical applications of lncRNAs in osteoporosis.

9.
J Immunol Res ; 2020: 1924379, 2020.
Article in English | MEDLINE | ID: mdl-32411789

ABSTRACT

Chimeric antigen receptor- (CAR-) T cell therapy is one of the most recent innovative immunotherapies and is rapidly evolving. Like other technologies, CAR-T cell therapy has undergone a long development process, and persistent explorations of the actions of the intracellular signaling domain and make several improvements have led to the superior efficacy when anti-CD19 CAR-T cell treatments in B cell cancers. At present, CAR-T cell therapy is developing rapidly, and many clinical trials have been established on a global scale, which has great commercial potential. This review mainly describes the toxicity of CAR-T cell therapy and the challenges of CAR-T cells in the treatment of solid tumors, and looks forward to future development and opportunities for immunotherapy and reviews major breakthroughs in CAR-T cell therapy.


Subject(s)
Cytokine Release Syndrome/immunology , Immunotherapy, Adoptive/adverse effects , Neoplasms/therapy , Neurotoxicity Syndromes/immunology , Receptors, Chimeric Antigen/immunology , Antigens, CD19/immunology , Antigens, CD19/metabolism , Cytokine Release Syndrome/prevention & control , Humans , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/trends , Neoplasms/immunology , Neurotoxicity Syndromes/prevention & control , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Treatment Outcome , Tumor Escape , Tumor Microenvironment/immunology
10.
J Biochem ; 168(3): 265-271, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32302384

ABSTRACT

As a research hotspot in recent years, bone mesenchymal stem cells (BMSCs) play an important role in the process of a variety of human diseases, including cancers. However, in osteosarcoma, the role of BMSCs and their communication with tumour cells are not clear. In this study, we validated the communication of osteosarcoma (OS) cells with BMSCs. The results showed that the conditioned medium of osteosarcoma cell line U2OS (U2OS-CM) induces the carcinoma-associated fibroblasts (CAFs)-like transformation of BMSCs and promotes the proliferation, migration and invasion of BMSCs. Mechanistically, treatment of human bone mesenchymal stem cells (hBMSCs) with U2OS-CM results in a significant increase in the IL-6 expression and phosphorylation of STAT3. Furthermore, blockade of the IL-6/STAT3 signalling in hBMSCs rescues the transformation of CAF phenotype induced by U2OS-CM. And, human IL-6 can directly increase the expression of the CAF marker genes in hMSCs. Meanwhile, IL-6/STAT3 signalling involves in promoting effects of U2OS-CM on the proliferation, migration and invasion of BMSCs. In summary, our results suggest that BMSCs communicate with OS cells through IL-6/STAT3 signalling and play an important role in the progress of osteosarcoma.


Subject(s)
Bone Marrow Cells/metabolism , Bone Neoplasms/metabolism , Fibroblasts/metabolism , Interleukin-6/metabolism , Mesenchymal Stem Cells/metabolism , Osteosarcoma/metabolism , STAT3 Transcription Factor/metabolism , Bone Marrow Cells/drug effects , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Humans , Interleukin-6/genetics , Male , Mesenchymal Stem Cells/drug effects , Osteosarcoma/pathology , Phenotype , Phosphorylation/drug effects , Signal Transduction/drug effects , Tissue Donors , Transfection
11.
Am J Transl Res ; 12(2): 332-342, 2020.
Article in English | MEDLINE | ID: mdl-32194887

ABSTRACT

Circulating tumor cells (CTCs) are cells that are shed from the primary tumor and circulate in the blood, and their metastasis and formation of a secondary tumor are closely associated with cancer-related death. Therefore, regulating tumor metastasis through CTCs can be a novel strategy to fight cancer. It has been demonstrated that CTCs can reflect the profile of the primary tumor and provide valuable information about intratumoral heterogeneity and their evolution over time. Moreover, the revelation of the relationship between metastasis and CTCs suggests that CTC regulation represents a promising novel anticancer strategy. Above all, at the molecular level, genetic analysis might be vital in the new era of gene-targeted cancer therapies and contribute to personalized anti-metastasis tumor treatments. In this review, we will focus on the biological significance of CTCs in the peripheral blood and discuss their potential clinical implications in cancer management.

13.
Am J Transl Res ; 12(12): 7693-7701, 2020.
Article in English | MEDLINE | ID: mdl-33437354

ABSTRACT

There are more than 100 sarcoma subtypes, each of which is uncommon and challenging to diagnose. Most patients with locally advanced and unresectable sarcomas are still treated with cytotoxic chemotherapy and have low long-term survival. Therefore, novel therapeutic methods are needed to improve the prognosis of patients with sarcomas. Immunotherapy is increasingly recognized as have an essential role in the treatment of malignant tumors. Emerging strategies, such as immune checkpoint inhibitors, vaccines, and adoptive cell therapies have been investigated for the treatment of sarcomas. Advances in these immunotherapies have provided a better understanding of how immuno-oncology can be best applied to the treatment of sarcomas, including their potential as adjuvant therapies in combination strategies. In this review, we discuss the immune microenvironment and how it relates to immunoresponsiveness, focusing on the advances in immunotherapy (immune checkpoint inhibitors, vaccines and adoptive cell therapies), the use of which will hopefully lead to improved outcomes for patients with sarcomas.

14.
J Cell Mol Med ; 23(8): 4854-4865, 2019 08.
Article in English | MEDLINE | ID: mdl-31210425

ABSTRACT

In the tumour microenvironment (TME), immunogenic cell death (ICD) plays a major role in stimulating the dysfunctional antitumour immune system. Chronic exposure of damage-associated molecular patterns (DAMPs) attracts receptors and ligands on dendritic cells (DCs) and activates immature DCs to transition to a mature phenotype, which promotes the processing of phagocytic cargo in DCs and accelerates the engulfment of antigenic components by DCs. Consequently, via antigen presentation, DCs stimulate specific T cell responses that kill more cancer cells. The induction of ICD eventually results in long-lasting protective antitumour immunity. Through the exploration of ICD inducers, recent studies have shown that there are many novel modalities with the ability to induce immunogenic cancer cell death. In this review, we mainly discussed and summarized the emerging methods for inducing immunogenic cancer cell death. Concepts and molecular mechanisms relevant to antitumour effects of ICD are also briefly discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Dendritic Cells/immunology , Immunogenic Cell Death/genetics , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Animals , Antineoplastic Agents/pharmacology , Calreticulin/genetics , Calreticulin/metabolism , Combined Modality Therapy , Endoplasmic Reticulum Stress/immunology , Humans , Immunotherapy , Mitochondrial Membranes/immunology , Mitochondrial Membranes/metabolism , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Phototherapy , Tumor Microenvironment/genetics
15.
Adv Sci (Weinh) ; 6(8): 1801233, 2019 Apr 17.
Article in English | MEDLINE | ID: mdl-31016106

ABSTRACT

Fullerene C60 nanocrystals (nano-C60) possess various attractive bioactivities, including autophagy induction and calcium/calmodulin-dependent protein kinase IIα (CaMKIIα) activation. CaMKIIα is a multifunctional protein kinase involved in many cellular processes including tumor progression; however, the biological effects of CaMKIIα activity modulated by nano-C60 in tumors have not been reported, and the relationship between CaMKIIα activity and autophagic degradation remains unclear. Herein, nano-C60 is demonstrated to elicit reactive oxygen species (ROS)-dependent cytotoxicity and persistent activation of CaMKIIα in osteosarcoma (OS) cells. CaMKIIα activation, in turn, produces a protective effect against cytotoxicity from nano-C60 itself. Inhibition of CaMKIIα activity by either the chemical inhibitor KN-93 or CaMKIIα knockdown dramatically promotes the anti-OS effect of nano-C60. Moreover, inhibition of CaMKIIα activity causes lysosomal alkalinization and enlargement, and impairs the degradation function of lysosomes, leading to autophagosome accumulation. Importantly, excessive autophagosome accumulation and autophagic degradation blocking are shown to play an important role in KN-93-enhanced-OS cell death. The synergistic anti-OS efficacy of KN-93 and nano-C60 is further revealed in an OS-xenografted murine model. The results demonstrate that CaMKIIα inhibition, along with the suppression of autophagic degradation, presents a promising strategy for improving the antitumor efficacy of nano-C60.

16.
Cancer Sci ; 110(5): 1746-1759, 2019 May.
Article in English | MEDLINE | ID: mdl-30907478

ABSTRACT

Osteosarcoma is the most common primary malignant bone tumor. Raddeanin A (RA) is an active oleanane-type triterpenoid saponin extracted from the traditional Chinese herb Anemone raddeana Regel that exerts antitumor activity against several cancer types. However, the effect of RA on osteosarcoma remains unclear. In the present study, we showed that RA inhibited proliferation and induced apoptosis of osteosarcoma cells in a dose- and time-dependent way in vitro and in vivo. RA treatment resulted in excessive reactive oxygen species (ROS) generation and JNK and ERK1/2 activation. Apoptosis induction was evaluated by the activation of caspase-3, caspase-8, and caspase-9 and poly-ADP ribose polymerase (PARP) cleavage. RA-induced cell death was significantly restored by the ROS scavenger glutathione (GSH), the pharmacological inhibitor of JNK SP600125, or specific JNK knockdown by shRNA. Additionally, signal transducer and activator of transcription 3 (STAT3) activation was suppressed by RA in human osteosarcoma, and this suppression was restored by GSH, SP600125, and JNK-shRNA. Further investigation showed that STAT3 phosphorylation was increased after JNK knockdown. In a tibial xenograft tumor model, RA induced osteosarcoma apoptosis and notably inhibited tumor growth. Taken together, our results show that RA suppresses proliferation and induces apoptosis by modulating the JNK/c-Jun and STAT3 signaling pathways in human osteosarcoma. Therefore, RA may be a promising candidate antitumor drug for osteosarcoma intervention.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Bone Neoplasms/drug therapy , Osteosarcoma/drug therapy , STAT3 Transcription Factor/metabolism , Saponins/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Osteosarcoma/metabolism , Reactive Oxygen Species/metabolism , Saponins/pharmacology , Time Factors , Xenograft Model Antitumor Assays
17.
Int J Cancer ; 145(4): 979-993, 2019 08 15.
Article in English | MEDLINE | ID: mdl-30719715

ABSTRACT

Osteosarcoma is the most common primary malignant bone tumor in children and adolescents, with highly aggressive behavior and early systemic metastasis. The survival rates for osteosarcoma remain unchanged over the past two decades. Studies aiming to find new or alternative therapies for patients with refractory osteosarcoma are urgently needed. Anlotinib, a novel multi-targeted tyrosine kinase inhibitor (TKI), has exhibited encouraging clinical activity in NSLCC and soft tissue sarcoma, whereas its effect on osteosarcoma has not been studied. In our study, we investigated the anti-tumor activity and underlying mechanism of anlotinib in osteosarcoma. Various in vitro and in vivo models of human osteosarcoma were used to determine the anti-proliferative, anti-angiogenesis and anti-metastasis efficacy of anlotinib. Our results showed that anlotinib suppressed tumor growth and increased the chemo-sensitivity of osteosarcoma. In addition, anlotinib inhibited migration and invasion in osteosarcoma cells. Furthermore, in order to explore the anti-tumor mechanism of anlotinib, phospho-RTK antibody arrays were performed. These analyses confirmed that anlotinib suppressed the phosphorylation of MET, VEGFR2 and the downstream signaling pathway activation. Moreover, we demonstrated that anlotinib blocked hepatocyte growth factor (HGF)-induced cell migration, invasion and VEGF-induced angiogenesis. Notably, a 143B-Luc orthotopic osteosarcoma model further showed that anlotinib significantly inhibited growth and lung metastasis of implanted tumor cells. Our preclinical work indicates that anlotinib acts as a novel inhibitor of VEGFR2 and MET that blocks tumorigenesis in osteosarcoma, which could be translated into future clinical trials.


Subject(s)
Bone Neoplasms/drug therapy , Indoles/pharmacology , Osteosarcoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Quinolines/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Hepatocyte Growth Factor/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis/drug therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Osteosarcoma/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects
18.
Int J Biol Sci ; 15(3): 668-679, 2019.
Article in English | MEDLINE | ID: mdl-30745853

ABSTRACT

Osteosarcoma (OS) is the most common primary bone malignancy in adolescents. One major obstacle for current OS treatment is drug-resistance. Raddeanin A (RA), an oleanane-type triterpenoid saponin, exerts anti-tumor effects in several tumor models, but the effect of RA in human drug-resistant OS remained to be elucidated. In the present study, we investigated the anti-tumor effects of RA in both drug-sensitive and drug-resistant OS cells and its underlying mechanism. RA inhibited cell proliferation and colony formation and induced apoptotic cell death in a dose-dependent manner in both drug-sensitive and drug-resistant cells. Moreover, RA exposure resulted in the inhibition of interleukin-6 (IL-6)-induced JAK2/STAT3 signaling pathway activation and target gene expression in both drug-sensitive and drug-resistant cells. Meanwhile, we observed significantly increased MDR1 and STAT3 expression in drug-resistant OS cells compared with parental cells. STAT3 overexpression promoted chemo-resistance and MDR1 protein expression in both drug-sensitive OS cells and drug-resistant OS cells, while inhibiting STAT3 with siRNA sensitized OS cells to doxorubicin treatment. In addition, RA synergistically increased doxorubicin toxicity by increasing its cellular uptake, ablating efflux and downregulating MDR1 in drug-resistant cells with attenuation of STAT3 Phosphorylation. Finally, RA suppressed in vivo tumor growth and induced apoptosis in nude mouse using drug-resistant OS tibia orthotopic model. Taken together, RA is a promising potential therapeutic for the treatment of doxorubicin resistance in OS.


Subject(s)
Osteosarcoma/chemically induced , Osteosarcoma/metabolism , STAT3 Transcription Factor/metabolism , Saponins/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/genetics , Female , Flow Cytometry , Humans , In Situ Nick-End Labeling , Interleukin-6/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction
19.
J Exp Clin Cancer Res ; 38(1): 44, 2019 Jan 31.
Article in English | MEDLINE | ID: mdl-30704503

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is a common malignant cancer in children and adolescents and has a cure rate that has not improved in the last two decades. CYT997 (lexibulin) is a novel potent microtubule-targeting agent with various anticancer activities, such as proliferation inhibition, vascular disruption, and cell cycle arrest and apoptosis induction, in multiple cancers. However, the direct cytotoxic mechanisms of CYT997 have not yet been fully characterized. METHODS: We evaluated apoptosis and autophagy in human osteosarcomas after treatment with CYT997 and investigated the underlying mechanisms. To explore relationships, we used the reactive oxygen species (ROS) scavenger N-acetyl cysteine (NAC), PERK inhibitor GSK2606414, ERO1 inhibitor EN460 and mitochondrial targeted protection peptide elamipretide. BALB/c-nu mice were inoculated with 143B tumor cells to investigate the in vivo effect of CYT997. RESULTS: We explored the efficacy and mechanism of CYT997 in osteosarcoma (OS) in vitro and in vivo and demonstrated that CYT997 potently suppresses cell viability and induces apoptosis and autophagy. CYT997 triggered production of ROS and exerted lethal effects via endoplasmic reticulum (ER) stress in OS cells. NAC attenuated these effects. The PERK inhibitor GSK2606414, which can block the ER stress pathway, reduced ROS production and enhanced cell viability. Moreover, activation of ERO1 in the ER stress pathway was responsible for inducing ROS production. ROS produced by the mitochondrial pathway also aggravate ER stress. Protection of mitochondria can reduce apoptosis and autophagy. Finally, CYT997 prominently reduced tumor growth in vivo. CONCLUSIONS: This study suggests that CYT997 induces apoptosis and autophagy in OS cells by triggering mutually enhanced ER stress and ROS and may thus be a promising agent against OS.


Subject(s)
Apoptosis/drug effects , Autophagy , Bone Neoplasms/pathology , Endoplasmic Reticulum Stress/drug effects , Osteosarcoma/pathology , Pyridines/pharmacology , Pyrimidines/pharmacology , Reactive Oxygen Species/metabolism , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
J Cell Mol Med ; 23(4): 2280-2292, 2019 04.
Article in English | MEDLINE | ID: mdl-30724027

ABSTRACT

Although the application of multiple chemotherapy brought revolutionary changes to improve overall survival of osteosarcoma patients, the existence of multidrug resistance (MDR) has become a great challenge for successful osteosarcoma treatment in recent decades. Substantial studies have revealed various underlying mechanisms of MDR in cancers. As for osteosarcoma, evidence has highlighted that microRNAs (miRNAs) can mediate in the processes of DNA damage response, apoptosis avoidance, autophagy induction, activation of cancer stem cells, and signal transduction. Besides, these drug resistance-related miRNAs showed much promise for serving as candidates for predictive biomarkers of poor outcomes and shorter survival time, and therapeutic targets to reverse drug resistance and overcome treatment refractoriness. This review aims to demonstrate the potential molecular mechanisms of miRNAs-regulated drug resistance in osteosarcoma, and provide insight in translating basic evidence into therapeutic strategies.


Subject(s)
Drug Resistance, Neoplasm/genetics , MicroRNAs/genetics , Osteosarcoma/drug therapy , Translational Research, Biomedical , Apoptosis/genetics , Autophagy/genetics , DNA Damage/drug effects , Humans , MicroRNAs/therapeutic use , Osteosarcoma/genetics , Osteosarcoma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...