Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Mol Cancer ; 23(1): 85, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678233

ABSTRACT

Nuclear condensates have been shown to regulate cell fate control, but its role in oncogenic transformation remains largely unknown. Here we show acquisition of oncogenic potential by nuclear condensate remodeling. The proto-oncogene SS18 and its oncogenic fusion SS18-SSX1 can both form condensates, but with drastically different properties and impact on 3D genome architecture. The oncogenic condensates, not wild type ones, readily exclude HDAC1 and 2 complexes, thus, allowing aberrant accumulation of H3K27ac on chromatin loci, leading to oncogenic expression of key target genes. These results provide the first case for condensate remodeling as a transforming event to generate oncogene and such condensates can be targeted for therapy. One sentence summary: Expulsion of HDACs complexes leads to oncogenic transformation.


Subject(s)
Histone Deacetylase 1 , Histone Deacetylase 2 , Proto-Oncogene Mas , Humans , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylase 2/genetics , Cell Nucleus/metabolism , Chromatin/metabolism , Chromatin/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Histones/metabolism , Animals
2.
Int Orthop ; 48(8): 2233-2241, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38847859

ABSTRACT

PURPOSE: To develop a novel classification of sagittal en bloc resection (SEBR) based on anatomical locations for thoracolumbar spine tumors and assess the clinical outcomes of this surgical procedure. METHODS: 31 patients with thoracolumbar tumours treated with SEBR were enrolled in this study. The individualized surgical strategy was adopted based on our surgical classification. Demographics, perioperative outcomes, complications and postoperative outcomes were assessed. RESULTS: Based on our surgical classifications, patients were divided into four types. All bony resection margins were negative, wide resection was achieved in 25 patients, marginal resection in four, and intralesional resection in two. 18 patients underwent anterior reconstruction. Complications were encountered in five patients, and instrumentation failure occurred in one patient. The median follow-up was 24 (range, 6-72) months and recurrence was found in only one patient. CONCLUSION: SEBR is a safe and effective surgical procedure for patients with thoracolumbar spinal tumours in specific anatomical locations. The proposed surgical classification covers all SEBR types and is easy to apply, it may assist surgical decision-making in patients with spinal tumours.


Subject(s)
Lumbar Vertebrae , Spinal Neoplasms , Thoracic Vertebrae , Humans , Thoracic Vertebrae/surgery , Male , Female , Middle Aged , Spinal Neoplasms/surgery , Spinal Neoplasms/pathology , Adult , Lumbar Vertebrae/surgery , Young Adult , Adolescent , Treatment Outcome , Aged , Retrospective Studies , Postoperative Complications/epidemiology , Postoperative Complications/etiology
3.
Eur Spine J ; 32(12): 4297-4305, 2023 12.
Article in English | MEDLINE | ID: mdl-37318598

ABSTRACT

PURPOSE: This study aimed to investigate whether short course of neoadjuvant denosumab treatment for spinal GCTB could (1) Induce radiological and histological response? (2) Facilitate en bloc resection? (3) Achieve satisfactory oncological and functional outcomes? METHODS: The clinical information of ten consecutive patients between 2018 and 2022 with spinal GCTB treated with short course of neoadjuvant denosumab (≤ 5 doses) and en bloc spondylectomy was retrospectively reviewed. The radiological and histological response, operative data, oncological and functional outcomes were analyzed. RESULTS: The mean doses of neoadjuvant denosumab were 4.2 (range 3-5 doses). After neoadjuvant denosumab, there were 9 cases showing new ossification and 5 cases with reappearance of cortical integrity. The values of Hounsfield units (HU) of the soft tissue component were increased by > 50% in 7 cases. The signal intensity (SI) ratios of tumor/muscle in T2WI of plain MRI were decreased by > 10% in 60% of the cases. Shrinkage of soft tissue mass by > 10% was observed in 4 cases. The mean duration of operation was 575 ± 174 min, and the mean estimated blood loss (EBL) was 2790 ± 1934 ml. No obvious adhesion to dura mater or major vessels was encounter intraoperatively. There is no tumor collapse or breakage during surgery. Multinucleated giant cells were decreased in 6 cases (60%) with the remaining 4 cases showing absence of multinucleated giant cells. Mononuclear stromal cells existed in most of the cases (8 cases, 80%). New bone formation was noticed in 8 cases (80%). No patient had a worsening of neurologic function after surgery. No tumor recurrence was noticed within the mean follow-up of 24 ± 20 months. CONCLUSION: Short-term neoadjuvant denosumab could yield radiological and histological responses and might facilitate en bloc spondylectomy by hardening the tumor and causing less adhesion to segmental vessels, major vessels and nerve roots, which was beneficial to achieve the optimal oncological and functional outcomes.


Subject(s)
Bone Density Conservation Agents , Bone Neoplasms , Giant Cell Tumor of Bone , Humans , Denosumab/adverse effects , Bone Density Conservation Agents/therapeutic use , Neoadjuvant Therapy , Treatment Outcome , Giant Cell Tumor of Bone/diagnostic imaging , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/surgery , Retrospective Studies , Neoplasm Recurrence, Local/surgery , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/drug therapy , Bone Neoplasms/surgery
4.
World J Surg Oncol ; 21(1): 385, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38097982

ABSTRACT

BACKGROUND: The outcomes of patients with tumors of the thoracolumbar spine treated with en bloc resection (EBR) using three-dimensional (3D)-printed endoprostheses are underreported. METHODS: We retrospectively evaluated patients with thoracolumbar tumors who underwent surgery at our institution. Logistic regression analysis was performed to identify the potential risk factors for surgical complications. Nomograms to predict complications were constructed and validated. RESULTS: A total of 53 patients with spinal tumors underwent EBR at our hospital; of these, 2 were lost to follow-up, 45 underwent total en bloc spondylectomy, and 6 were treated with sagittal en bloc spondylectomy. The anterior reconstruction materials included a customized 3D-printed artificial vertebral body (AVB) in 10 cases and an off-the-shelf 3D-printed AVB in 41 cases, and prosthesis mismatch occurred in 2 patients reconstructed with the off-the-shelf 3D-printed AVB. The median follow-up period was 21 months (range, 7-57 months). Three patients experienced local recurrence, and 5 patients died at the final follow-up. A total of 50 perioperative complications were encountered in 29 patients, including 25 major and 25 minor complications. Instrumentation failure occurred in 1 patient, and no prosthesis subsidence was observed. Using a combined surgical approach was a dependent predictor of overall complications, while Karnofsky performance status score, lumbar spine lesion, and intraoperative blood loss ≥ 2000 mL were predictors of major complications. Nomograms for the overall and major complications were constructed using these factors, with C-indices of 0.850 and 0.891, respectively. CONCLUSIONS: EBR is essential for the management of thoracolumbar tumors; however, EBR has a steep learning curve and a high complication rate. A 3D-printed AVB is an effective and feasible reconstruction option for patients treated with EBR.


Subject(s)
Spinal Neoplasms , Vertebral Body , Humans , Vertebral Body/pathology , Retrospective Studies , Spinal Neoplasms/surgery , Spinal Neoplasms/pathology , Treatment Outcome , Printing, Three-Dimensional
5.
BMC Musculoskelet Disord ; 23(1): 273, 2022 Mar 22.
Article in English | MEDLINE | ID: mdl-35317753

ABSTRACT

BACKGROUND: Minimally invasive separation surgery (MISS) is a safe and effective surgical technique, the current optimal treatment for spinal metastases. However, the learning curve for this technique has not been analyzed. This study aimed to define and analyze the surgical learning curve of MISS for the treatment of spinal metastases with small incision and freehand pedicle screw fixation. METHODS: A continuous series of 62 patients with spinal metastases who underwent MISS were included. Each patient's operative data were accurately counted. The improvement of the patients' neurological function was followed up after surgery to evaluate the surgical treatment effect. Logarithmic curve-fit regression was used to analyze the surgical learning curve of MISS. The number of cases needed to achieve proficiency was analyzed. Based on this cut-off point, this series of cases was divided into the early phase and later phase groups. The influence of the time sequence of MISS on surgical data and surgical efficacy was analyzed. RESULTS: The operative time decreased gradually with the number of surgical cases increasing and stabilized after the 20th patient. There was no statistical difference in demographic characteristics and preoperative characteristics between the two groups. The mean operative time in the later phase group was about 39 min shorter than that in the early phase group (mean 227.95 vs. 189.02 min, P = 0.027). However, it did not affect other operative data or the surgical treatment effect. CONCLUSION: The learning curve of MISS for spinal metastases is not steep. With the increase of surgeons' experience, the operative time drops rapidly and stabilizes within a certain range. MISS can be safely and effectively performed at the beginning of a surgeon's caree.


Subject(s)
Pedicle Screws , Spinal Fusion , Spinal Neoplasms , Humans , Learning Curve , Lumbar Vertebrae/surgery , Spinal Fusion/methods , Spinal Neoplasms/diagnostic imaging , Spinal Neoplasms/surgery , Treatment Outcome
6.
Cancer Cell Int ; 21(1): 88, 2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33632212

ABSTRACT

BACKGROUND: Utrophin (UTRN), as a tumor suppressor gene, is involved in various cancer progression. The function of UTRN in the melanoma process and the related molecular mechanisms are still unclear. Herein, we studied the function of UTRN in melanoma growth and the relevant molecular mechanisms. METHODS: Using the GEO database and UCSC Xena project, we compared the expression of UTRN in non-cancerous and melanoma tissues. Immunohistochemistry (IHC) staining, qRT-PCR and Western Blot (WB) were performed to evaluate UTRN expression in clinical samples. A total of 447 cases with UTRN expression data, patient characteristics and survival data were extracted from TCGA database and analyzed. After stable transduction and single cell cloning, the proliferation ability of A375 human melanoma cells was analyzed by Cell Counting Kit­8 (CCK) and 5­ethynyl­2'­deoxyuridine (EdU) incorporation assays. GSEA was performed to predict the mechanism by which UTRN regulated melanoma growth. Then WB analysis was used to assess the protein expression levels of pathway signaling in overexpression (EXP) melanoma cells. Epac activator 8-pCPT-2'-O-Me-cAMP was then used to evaluate the proliferation ability by activation of p38 and JNK/c-Jun signaling pathways. RESULTS: Data from GEO and UCSC Xena project indicated that UTRN expression was decreased in melanoma. Experiment on clinical samples further confirmed our finding. TCGA results showed that a reduced expression of UTRN in 447 melanoma samples was associated with advanced clinical characteristics (T stage, Clark level, ulceration), shorter survival time and poorer prognosis. In addition, up-regulated UTRN expression inhibited melanoma cell proliferation when compared to control group. MAPK signaling pathway was presented in both KEGG and BioCarta databases by using GSEA tool. WB results confirmed the down-regulated expression of p38, JNK1 and c-Jun in EXP group when compared to control group. Epac activator 8-pCPT-2'-O-Me-cAMP treatment could partially rescue proliferation of tumor cells. CONCLUSION: We have demonstrated that reduced UTRN predicted poorer prognosis and UTRN inhibited melanoma growth via p38 and JNK1/c-Jun pathways. Therefore, UTRN could serve as a tumor suppressor and novel prognostic biomarker for melanoma patients.

7.
Cancer Sci ; 111(6): 1899-1909, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32232912

ABSTRACT

Tumor-infiltrating immune cells play a crucial role in tumor progression and response to treatment. However, the limited studies on infiltrating immune cells have shown inconsistent and even controversial results for osteosarcoma (OS). In addition, the dynamic changes of infiltrating immune cells after neoadjuvant chemotherapy are largely unknown. We downloaded the RNA expression matrix and clinical information of 80 OS patients from the TARGET database. CIBERSORT was used to evaluate the proportion of 22 immune cell types in patients based on gene expression data. M2 macrophages were found to be the most abundant immune cell type and were associated with improved survival in OS. Another cohort of pretreated OS samples was evaluated by immunohistochemistry to validate the results from CIBERSORT analysis. Matched biopsy and surgical samples from 27 patients were collected to investigate the dynamic change of immune cells and factors before and after neoadjuvant chemotherapy. Neoadjuvant chemotherapy was associated with increased densities of CD3+ T cells, CD8+ T cells, Ki67 + CD8+ T cells and PD-L1+ immune cells. Moreover, HLA-DR-CD33+ myeloid-derived suppressive cells (MDSC) were decreased after treatment. We determined that the application of chemotherapy may activate the local immune status and convert OS into an immune "hot" tumor. These findings provide rationale for investigating the schedule of immunotherapy treatment in OS patients in future clinical trials.


Subject(s)
Bone Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Osteosarcoma/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Adolescent , Adult , Antineoplastic Agents, Immunological/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Chemotherapy, Adjuvant/methods , Child , Female , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Macrophages/drug effects , Macrophages/immunology , Male , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Neoadjuvant Therapy/methods , Osteosarcoma/drug therapy , Osteosarcoma/pathology
8.
Int J Cancer ; 143(5): 1134-1142, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29569716

ABSTRACT

Osteosarcoma is a primary malignant bone tumor that has a high potential to metastasize to lungs. Little is known about the mechanisms underlying the dissemination of OS cancer cells to lungs. We performed whole exome sequencing of 13 OS primary tumors, with matched lung metastases and normal tissues. Phylogenetic analyses revealed that lung metastatic tumors often harbor clones that are nonexistent or rare in the matched primary OS tumors. Spatially and temporally separated lung metastases were from parallel seeding events with a polyphyletic pattern. Loss of TP53 or RB1 is among the early events during OS tumorigenesis, while loss of PTEN is involved at the later stages associated with lung metastases. Finally, KEAP1 was identified as a novel biomarker for increased metastatic risk. Patients whose primary tumors harbored KEAP1 amplification have significantly poorer lung-metastasis free survival. This finding was validated in two independent datasets. Further, in vitro experiments exhibited that KEAP1 depletion suppressed the invasion of OS cells. Our findings uncover the patterns of clonal evolution during OS progression and highlight KEAP1 as a novel candidate associated with the risk of lung metastasis in OS patients.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/pathology , Clonal Evolution , Kelch-Like ECH-Associated Protein 1/genetics , Lung Neoplasms/secondary , Mutation , Osteosarcoma/pathology , Bone Neoplasms/genetics , Disease Progression , Gene Amplification , Humans , Lung Neoplasms/genetics , Osteosarcoma/genetics , PTEN Phosphohydrolase/genetics , Prognosis , Survival Rate , Tumor Suppressor Protein p53/genetics , Exome Sequencing
9.
J Plast Reconstr Aesthet Surg ; 89: 105-116, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38160589

ABSTRACT

The volume retention of breast autologous fat grafting is unpredictable, and the volume retention rate and related influencing factors have not been systematically reviewed. Therefore, this systematic review and meta-analysis aimed at evaluating the volume retention rate and related influencing factors of breast autologous fat grafting, which is for reconstructive or esthetic purposes. Literature search was conducted using the PubMed, Embase, Cochrane Library, and Web of Science databases from inception of study to December 2022. Sensitivity analysis was performed for all outcomes. Begg's test was performed to test publication bias. Subgroup analysis was performed based on population, method of fat preparation, method of volume measurement, number of fat grafting, and injected fat grafting volume. A total of 25 studies were included in this systematic review and meta-analysis. The follow-up time ranged from 3 to 36 months. Results showed that the pooled volume retention rate at the latest follow-up point was 54% [95% confidence interval (CI): 48.5%-59.5%]. Based on the fat preparation methods, the pooled rate was 51.5% (95%CI: 41.5%-61.5%) for centrifugation, which was higher than that for sedimentation [38.7% (95%CI: 30.9%-46.5%)]. In addition, the enrichment of stromal vascular fraction obtained after centrifugation was found to be associated with higher volume retention rate (weighted mean difference: 17.36, 95%CI: 8.84-25.87). Our findings may provide guidance for evaluating the volume retention rate of breast autologous fat grafting in clinical settings. Further studies are needed to verify our findings.


Subject(s)
Adipose Tissue , Mammaplasty , Transplantation, Autologous , Humans , Adipose Tissue/transplantation , Mammaplasty/methods , Female , Graft Survival , Breast/surgery
10.
Bone Jt Open ; 5(4): 317-323, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38631693

ABSTRACT

Aims: The aim of this study was to investigate the safety and efficacy of 3D-printed modular prostheses in patients who underwent joint-sparing limb salvage surgery (JSLSS) for malignant femoral diaphyseal bone tumours. Methods: We retrospectively reviewed 17 patients (13 males and four females) with femoral diaphyseal tumours who underwent JSLSS in our hospital. Results: In all, 17 patients with locally aggressive bone tumours (Enneking stage IIB) located in the femoral shaft underwent JSLSS and reconstruction with 3D-printed modular prostheses between January 2020 and June 2022. The median surgical time was 153 minutes (interquartile range (IQR) 117 to 248), and the median estimated blood loss was 200ml (IQR 125 to 400). Osteosarcoma was the most common pathological type (n = 12; 70.6%). The mean osteotomy length was 197.53 mm (SD 12.34), and the median follow-up was 25 months (IQR 19 to 38). Two patients experienced local recurrence and three developed distant metastases. Postoperative complications included wound infection in one patient and screw loosening in another, both of which were treated successfully with revision surgery. The median Musculoskeletal Tumor Society score at the final follow-up was 28 (IQR 27 to 28). Conclusion: The 3D-printed modular prosthesis is a reliable and feasible reconstruction option for patients with malignant femoral diaphyseal tumours. It helps to improve the limb salvage rate, restore limb function, and achieve better short-term effectiveness.

11.
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
12.
Cell Rep ; 43(2): 113751, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38341855

ABSTRACT

The premetastatic niche (PMN) contributes to lung-specific metastatic tropism in osteosarcoma. However, the crosstalk between primary tumor cells and lung stromal cells is not clearly defined. Here, we dissect the composition of immune cells in the lung PMN and identify granulocytic myeloid-derived suppressor cell (gMDSC) infiltration as positively associated with immunosuppressive PMN formation and tumor cell colonization. Osteosarcoma-cell-derived extracellular vesicles (EVs) activate lung interstitial macrophages to initiate the influx of gMDSCs via secretion of the chemokine CXCL2. Proteomic profiling of EVs reveals that EV-packaged S100A11 stimulates the Janus kinase 2/signal transducer and activator of transcription 3 signaling pathway in macrophages by interacting with USP9X. High level of S100A11 expression or circulating gMDSCs correlates with the presentation of lung metastasis and poor prognosis in osteosarcoma patients. In summary, we identify a key role of tumor-derived EVs in lung PMN formation, providing potential strategies for monitoring or preventing lung metastasis in osteosarcoma.


Subject(s)
Bone Neoplasms , Extracellular Vesicles , Lung Neoplasms , Osteosarcoma , Humans , Proteomics , S100 Proteins , Ubiquitin Thiolesterase
13.
Mol Cancer Res ; 22(4): 402-414, 2024 04 02.
Article in English | MEDLINE | ID: mdl-38226993

ABSTRACT

Bone metastasis (BM) is one of the most common complications of advanced cancer. Immunotherapy for bone metastasis of lung cancer (LCBM) is not so promising and the immune mechanisms are still unknown. Here, we utilized a model of BM by injecting cancer cells through caudal artery (CA) to screen out a highly bone metastatic derivative (LLC1-BM3) from a murine lung cancer cell line LLC1. Mass spectrometry-based proteomics was performed in LLC1-parental and LLC1-BM3 cells. Combining with prognostic survival information from patients with lung cancer, we identified serpin B9 (SB9) as a key factor in BM. Molecular characterization showed that SB9 overexpression was associated with poor prognosis and high bone metastatic burden in lung cancer. Moreover, SB9 could increase the ability of lung cancer cells to metastasize to the bone. The mechanistic studies revealed that tumor-derived SB9 promoted BM through an immune cell-dependent way by inactivating granzyme B, manifesting with the decreased infiltration of cytotoxic T cells and increased expression level of exhausted markers. A specific SB9-targeting inhibitor [1,3-benzoxazole-6-carboxylic acid (BTCA)] significantly suppressed LCBM in the CA mouse model. This study reveals that SB9 may serve as a therapeutic target and potential prognostic marker for patients with LCBM. IMPLICATIONS: SB9 as a therapeutic target for LCBM.


Subject(s)
Bone Neoplasms , Lung Neoplasms , Serpins , Humans , Mice , Animals , Lung Neoplasms/pathology , Serpins/genetics , Serpins/metabolism , Proteomics , Cell Line , Bone Neoplasms/genetics
14.
Cancer Res ; 84(7): 994-1012, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38295227

ABSTRACT

Cooperation between primary malignant cells and stromal cells can mediate the establishment of lung metastatic niches. Here, we characterized the landscape of cell populations in the tumor microenvironment in treatment-naïve osteosarcoma using single-cell RNA sequencing and identified a stem cell-like cluster with tumor cell-initiating properties and prometastatic traits. CXCL14 was specifically enriched in the stem cell-like cluster and was also significantly upregulated in lung metastases compared with primary tumors. CXCL14 induced stromal reprogramming and evoked a malignant phenotype in fibroblasts to form a supportive lung metastatic niche. Binding of CXCL14 to heterodimeric integrin α11ß1 on fibroblasts activated actomyosin contractility and matrix remodeling properties. CXCL14-stimulated fibroblasts produced TGFß and increased osteosarcoma invasion and migration. mAbs targeting the CXCL14-integrin α11ß1 axis inhibited fibroblast TGFß production, enhanced CD8+ T cell-mediated antitumor immunity, and suppressed osteosarcoma lung metastasis. Taken together, these findings identify cross-talk between osteosarcoma cells and fibroblasts that promotes metastasis and demonstrate that targeting the CXCL14-integrin α11ß1 axis is a potential strategy to inhibit osteosarcoma lung metastasis. SIGNIFICANCE: Cooperation between stem-like osteosarcoma cells and fibroblasts mediated by a CXCL14-integrin α11ß1 axis creates a tumor-supportive lung metastatic niche and represents a therapeutic target to suppress osteosarcoma metastasis.


Subject(s)
Chemokines, CXC , Integrins , Lung Neoplasms , Osteosarcoma , Tumor Microenvironment , Humans , Cell Line, Tumor , Chemokines, CXC/metabolism , Fibroblasts/metabolism , Integrins/metabolism , Lung/pathology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Osteosarcoma/pathology , Receptors, Collagen , Transforming Growth Factor beta/metabolism
15.
Spine (Phila Pa 1976) ; 46(1): 62-68, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-32925689

ABSTRACT

STUDY DESIGN: A retrospective study was conducted. OBJECTIVE: This study aims to compare the perioperative outcomes of minimal invasive spine surgery (MISS) and traditional open surgery (TOS) for thoracolumbar spine metastasis. SUMMARY OF BACKGROUND DATA: TOS for metastatic spinal tumors has many disadvantages, such as significant blood loss and high complication rate. MISS may change the treatment modality, but its safety and efficacy for spinal metastasis are lacking. METHODS: We retrospectively reviewed clinical data from 154 consecutive patients registered in our institute who underwent separation surgery for spinal metastases from January 2017 to December 2019. Forty-nine patients received MISS and 105 patients had TOS. The demographic and perioperative data were collected and compared between two approaches. RESULTS: There were no significant differences in baseline characteristics between the MISS and TOS group, except the sex (P = 0.04). The mean intraoperative blood loss in MISS group was lower than that in TOS group (748.57 vs. 950.48 mL, P = 0.039). The operative time was comparable between both groups (mean 213.45 vs. 221.03 minutes, P = 0.78). The postoperative drainage before discharge in MISS group was remarkably less than that in TOS group (mean 494.02 vs. 1099.10 mL, P = 0.0004). As compared to TOS group, patients in MISS group had lower complication rate, although the difference is not significant (9.52% vs. 6.12%, P = 0.55). The infection rate of MISS group was two-fold lower than that in the TOS group, although the difference is not significant (P = 0.43). The mean hospital stay of MISS group is 7.35 days, which is significantly shorter than TOS group (9.94 days, P = 0.0007). Patients in both groups exhibited similar postoperative neurological outcomes. CONCLUSION: MISS is a safe and effective technique that could be considered the optimal treatment for patients with spinal metastasis and myelopathy and thus is an excellent alternative in managing thoracolumbar spine metastasis. LEVEL OF EVIDENCE: 3.


Subject(s)
Minimally Invasive Surgical Procedures , Spinal Neoplasms/surgery , Adult , Aged , Blood Loss, Surgical , Female , Humans , Length of Stay , Lumbar Vertebrae/surgery , Male , Middle Aged , Operative Time , Retrospective Studies , Spinal Cord Diseases , Treatment Outcome
16.
Front Cell Dev Biol ; 9: 780951, 2021.
Article in English | MEDLINE | ID: mdl-35186946

ABSTRACT

Growing evidence indicates a link between retinoic acid (RA) metabolism and sarcoma progression or immunity in laboratory studies. However, a comprehensive analysis of RA abnormality in the sarcoma population is still lacking. Herein, we systematically analyzed the molecular features of 19 retinoic acid metabolism-related enzymes and sarcoma patients' clinical information based on TCGA/TARGET/GSE datasets. We identified two RA expression subtypes, which were related to distinct clinical survival outcomes and exhibited different biological features. Gene set enrichment analysis indicated a set of immune pathways were enriched in G1 while oncogenic pathways were enriched in G2. Immune cell infiltration analysis using the TIMER algorithm revealed more CD4+ and CD8+ T cell infiltration in G1 subgroups than in G2. Moreover, we generated a seven genes signature to predict the RA metabolism index based on the LASSO-penalized Cox regression model. Survival analysis demonstrated the significant prognostic differences between high- and low-risk groups among different bone and soft tissue datasets. A higher risk index was associated with less T cell CD8+ infiltration. The predictive ability of the RA risk score was validated in 71 bone or soft tissue sarcoma clinical samples. These results indicated that RA-based classification could distinguish sarcoma patients with different clinical outcomes and immune statuses, which may help to explore better treatment decision-making for sarcoma patients.

17.
Front Immunol ; 12: 623762, 2021.
Article in English | MEDLINE | ID: mdl-33959121

ABSTRACT

Osteosarcoma (OSA) is the most common bone malignancy and displays high heterogeneity of molecular phenotypes. This study aimed to characterize the molecular features of OSA by developing a classification system based on the gene expression profile of the tumor microenvironment. Integrative analysis was performed using specimens and clinical information for OSA patients from the TARGET program. Using a matrix factorization method, we identified two molecular subtypes significantly associated with prognosis, S1 (infiltration type) and S2 (escape type). Both subtypes displayed unique features of functional significance features and cellular infiltration characteristics. We determined that immune and stromal infiltrates were abundant in subtype S1 compare to that in subtype S2. Furthermore, higher expression of immune checkpoint PDCD1LG2 and HAVCR2 was associated with improved prognosis, while a preferable chemotherapeutic response was associated with FAP-positive fibroblasts in subtype S1. Alternatively, subtype S2 is characterized by a lack of effective cytotoxic responses and loss of major histocompatibility complex class I molecule expression. A gene classifier was ultimately generated to enable OSA classification and the results were confirmed using the GSE21257 validation set. Correlations between the percentage of fibroblasts and/or fibrosis and CD8+ cells, and their clinical responses to chemotherapy were assessed and verified based on 47 OSA primary tumors. This study established a new OSA classification system for stratifying OSA patient risk, thereby further defining the genetic diversity of OSA and allowing for improved efficiency of personalized therapy.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , CD8-Positive T-Lymphocytes/immunology , Cancer-Associated Fibroblasts/pathology , Gene Expression Profiling , Lymphocytes, Tumor-Infiltrating/immunology , Osteosarcoma/genetics , Transcriptome , Tumor Microenvironment , Adolescent , Adult , Biomarkers, Tumor/metabolism , Bone Neoplasms/immunology , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , CD8-Positive T-Lymphocytes/metabolism , Cancer-Associated Fibroblasts/metabolism , Child , Databases, Genetic , Female , Fibrosis , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Osteosarcoma/immunology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Phenotype , Predictive Value of Tests , Prognosis , Young Adult
18.
J Invest Dermatol ; 140(2): 395-403.e6, 2020 02.
Article in English | MEDLINE | ID: mdl-31401046

ABSTRACT

SHARPIN, as a tumor-associated gene, is involved in the metastatic process of many kinds of tumors. Herein, we studied the function of Shank-associated RH domain interacting protein (SHARPIN) in melanoma metastasis and the relevant molecular mechanisms. We found that SHARPIN expression was increased in melanoma tissues and activated the process of proliferation, migration, and invasion in vitro and in vivo, resulting in a poor prognosis of the disease. Functional analysis demonstrated that SHARPIN promoted melanoma migration and invasion by regulating Ras-associated protein-1(Rap1) and its downstream pathways, including p38 and JNK/c-Jun. Rap1 activator (8-pCPT-2'-O-Me-cAMP) and inhibitor (ESI-09 and farnesylthiosalicylic acid-amide) treatments could partially rescue invasion and migration of tumor cells. Additionally, SHARPIN expression in cell lines and public datasets also indicated that molecules other than BRAF and N-RAS may contribute to SHARPIN activation. In conclusion, our broad-in-depth work suggests that SHARPIN promotes melanoma development via p38 and JNK/c-Jun pathways through upregulation of Rap1 expression.


Subject(s)
Melanoma/pathology , Skin Neoplasms/pathology , Telomere-Binding Proteins/metabolism , Ubiquitins/metabolism , Adult , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Movement/drug effects , Datasets as Topic , Disease Progression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , MAP Kinase Signaling System/drug effects , Male , Melanoma/mortality , Mice , Middle Aged , Neoplasm Invasiveness/pathology , Prognosis , Shelterin Complex , Skin/pathology , Skin Neoplasms/mortality , Telomere-Binding Proteins/agonists , Telomere-Binding Proteins/antagonists & inhibitors , Ubiquitins/genetics , Up-Regulation , Xenograft Model Antitumor Assays
19.
Aging (Albany NY) ; 13(2): 2168-2183, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33316779

ABSTRACT

Tumor microenvironments are strongly related to tumor development, and immune-infiltrating cells and immune-related molecules are potential prognostic markers. However, the shortcomings of traditional measurement methods limit the accurate evaluation of various components in tumor microenvironments. With the rapid advancement of Next-Generation RNA Sequencing technology, dedicated and in-depth analyses of immune filtration within the tumor microenvironment has been achieved. In this study, we combined the bioinformatics analysis methods ESTIMATE, CIBERSORT, and ssGSEA to characterize the immune infiltration of sarcomas and to identify specific immunomodulators of different pathological subtypes. We further extracted a functional enrichment of significant immune-related genes related to improved prognosis, including NR1H3, VAMP5, GIMAP2, GBP2, HLA-E and CRIP1. Overall, the immune microenvironment is an important prognostic determinant of sarcomas and may be a potential resource for developing effective immunotherapy.


Subject(s)
Gene Expression Regulation, Neoplastic , Mutation , Sarcoma/genetics , Tumor Microenvironment/genetics , Biomarkers, Tumor/genetics , Carrier Proteins/genetics , Computational Biology , Female , GTP Phosphohydrolases/genetics , GTP-Binding Proteins/genetics , Histocompatibility Antigens Class I/genetics , Humans , LIM Domain Proteins/genetics , Liver X Receptors/genetics , Male , Membrane Proteins/genetics , Prognosis , R-SNARE Proteins/genetics , Sarcoma/immunology , Sarcoma/mortality , Tumor Microenvironment/immunology , HLA-E Antigens
20.
Front Oncol ; 10: 1198, 2020.
Article in English | MEDLINE | ID: mdl-32850346

ABSTRACT

Background: Osteosarcoma (OSA), the most common primary bone malignancy in children and adolescents, is prone to metastases and unfavorable prognosis. Owing to its strong genomic heterogeneity, traditional chemotherapy, or targeted immunotherapy has not effectively improved the related overall survival for decades. Since the landscape of the OSA tumor immune microenvironment is scarcely known, despite it playing a crucial role in predicting clinical outcomes and therapeutic efficacies, we aimed to elucidate its molecular characteristics. Methods: The immune signature of 101 OSA samples was explored using transcriptome profiling and clinical characteristics retrieved from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) program. Correlations between the prognostic immune markers and their clinical chemotherapy responses were assessed and verified based on 45 OSA primary tumors. Findings: We identified the heterogeneity underlying tumor immune signature in OSA, and found CD4+ T cells and macrophage markers CD4/IFNGR2/CD68 to be feasible prognostic factors, exerting significantly positive correlation with each other. Specifically, CSF1R, which plays an essential role in the regulation of proliferation and differentiation of macrophages, was found to be a specific signature associated with CD4/CD68, with improved OSA clinical outcomes. Interpretation: The immune landscape based on CD4/CD68/CSF1R gene signatures showed considerable promise for prognostic and therapeutic stratification in OSA patients. A specific immune signature for OSA, abundantly consisting of Th1-polarized CD4+ T cells and CSF1R-related CD68+ macrophages, may improve the predictive efficacy of chemotherapy and improve prognosis in patients with OSA.

SELECTION OF CITATIONS
SEARCH DETAIL