Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cells ; 13(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38891109

ABSTRACT

Chondrosarcoma (ChS), a malignant cartilage-producing tumor, is the second most frequently diagnosed osseous sarcoma after osteosarcoma. It represents a very heterogeneous group of malignant chemo- and radiation-resistant neoplasms, accounting for approximately 20% of all bone sarcomas. The majority of ChS patients have a good prognosis after a complete surgical resection, as these tumors grow slowly and rarely metastasize. Conversely, patients with inoperable disease, due to the tumor location, size, or metastases, represent a great clinical challenge. Despite several genetic and epigenetic alterations that have been described in distinct ChS subtypes, very few therapeutic options are currently available for ChS patients. Therefore, new prognostic factors for tumor progression as well as new treatment options have to be explored, especially for patients with unresectable or metastatic disease. Recent studies have shown that a correlation between immune infiltrate composition, tumor aggressiveness, and survival does exist in ChS patients. In addition, the intra-tumor microvessel density has been proven to be associated with aggressive clinical behavior and a high metastatic potential in ChS. This review will provide an insight into the ChS microenvironment, since immunotherapy and antiangiogenic agents are emerging as interesting therapeutic options for ChS patients.


Subject(s)
Chondrosarcoma , Tumor Microenvironment , Humans , Chondrosarcoma/pathology , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Immunotherapy , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/pharmacology
2.
Explor Target Antitumor Ther ; 4(3): 498-510, 2023.
Article in English | MEDLINE | ID: mdl-37455823

ABSTRACT

Soft tissue sarcomas (STSs) are rare, heterogeneous, and very often asymptomatic diseases. Their diagnosis is fundamental, as is the identification of the degree of malignancy, which may be high, medium, or low. The Italian Medical Oncology Association and European Society of Medical Oncology (ESMO) guidelines recommend magnetic resonance imaging (MRI) because the clinical examination is typically ineffective. The diagnosis of these rare diseases with artificial intelligence (AI) techniques presents reduced datasets and therefore less robust methods. However, the combination of AI techniques with radiomics may be a new angle in diagnosing rare diseases such as STSs. Results obtained are promising within the literature, not only for the performance but also for the explicability of the data. In fact, one can make tumor classification, site localization, and prediction of the risk of developing metastasis. Thanks to the synergy between computer scientists and radiologists, linking numerical features to radiological evidence with excellent performance could be a new step forward for the diagnosis of rare diseases.

3.
Cancers (Basel) ; 14(14)2022 Jul 14.
Article in English | MEDLINE | ID: mdl-35884474

ABSTRACT

The major histocompatibility complex (MHC) class I expression in cancer cells has a crucial impact on the outcome of T cell-mediated cancer immunotherapy. We now determined the HLA class I allelic variants and their expression in PD-L1-deficient and positive rare sarcoma tissues. Tumor tissues were HLA-I classified based on HLA-A and -B alleles, and for class II, the HLA-DR-B by Taqman genomic PCRs. The HLA-A24*:10-B73*:01 haplotype was the most common. A general down-regulation or deletion of HLA-B mRNA and HLA-A was observed, compared to HLA-DR-B. HLA-I was almost too low to be detectable by immunohistochemistry and 32% of grade III cases were positive to PD-L1. Functional cytotoxic assays co-culturing patient biopsies with autologous T cells were used to assess their ability to kill matched tumor cells. These results establish that deletion of HLA-I loci together with their down-regulation in individual patient restrict the autologous lymphocyte cytotoxic activity, even in the presence of the immune checkpoint blocking antibody, Nivolumab. Additionally, the proposed cytotoxic test suggests a strategy to assess the sensitivity of tumor cells to T cell-mediated attack at the level of the individual patient.

4.
Cell Death Dis ; 12(9): 797, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34404763

ABSTRACT

The tumor microenvironment modulates cancer growth. Extracellular vesicles (EVs) have been identified as key mediators of intercellular communication, but their role in tumor growth is largely unexplored. Here, we demonstrate that EVs from sarcoma patients promote neoangiogenesis via a purinergic X receptor 4 (P2XR4) -dependent mechanism in vitro and in vivo. Using a proteomic approach, we analyzed the protein content of plasma EVs and identified critical activated pathways in human umbilical vein endothelial cells (HUVECs) and human progenitor hematopoietic cells (CD34+). We then showed that vessel formation was due to rapid mitochondrial activation, intracellular Ca2+ mobilization, increased extracellular ATP, and trafficking of the lysosomal P2XR4 to the cell membrane, which is required for cell motility and formation of stable branching vascular networks. Cell membrane translocation of P2XR4 was induced by proteins and chemokines contained in EVs (e.g. Del-1 and SDF-1). Del-1 was found expressed in many EVs from sarcoma tumors and several tumor types. P2XR4 blockade reduced EVs-induced vessels in angioreactors, as well as intratumor vascularization in mouse xenografts. Together, these findings identify P2XR4 as a key mediator of EVs-induced tumor angiogenesis via a signaling mediated by mitochondria-lysosome-sensing response in endothelial cells, and indicate a novel target for therapeutic interventions.


Subject(s)
Cell-Derived Microparticles/metabolism , Lysosomes/metabolism , Neovascularization, Pathologic/metabolism , Receptors, Purinergic P2X4/metabolism , Sarcoma/blood supply , Sarcoma/pathology , Animals , Calcium/metabolism , Cell Movement , Cytosol/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mitochondria/metabolism , Retina/pathology , Sarcoma/blood , Signal Transduction , Viscosity
5.
Cancers (Basel) ; 13(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34209309

ABSTRACT

Myxoid liposarcoma (MLPS) is the second most common subtype of liposarcoma and has tendency to metastasize to soft tissues. To date, the mechanisms of invasion and metastasis of MLPS remain unclear, and new therapeutic strategies that improve patients' outcomes are expected. In this study, we analyzed by immunohistochemistry the immune cellular components and microvessel density in tumor tissues from patients affected by MLPS. In order to evaluate the effects of primary human MLPS cells on macrophage polarization and, in turn, the ability of macrophages to influence invasiveness of MLPS cells, non-contact and 3D organotypic co-cultures were set up. High grade MLPS tissues were found heavily vascularized, exhibited a CD3, CD4, and CD8 positive T lymphocyte-poor phenotype and were massively infiltrated by CD163 positive M2-like macrophages. Conversely, low grade MLPS tissues were infiltrated by a discrete amount of CD3, CD4, and CD8 positive T lymphocytes and a scarce amount of CD163 positive macrophages. Kaplan-Meier analysis revealed a shorter Progression Free Survival in MLPS patients whose tumor tissues were highly vascularized and heavily infiltrated by CD163 positive macrophages, indicating a clear-cut link between M2-like macrophage abundance and poor prognosis in patients. Moreover, we documented that, in co-culture, soluble factors produced by primary human MLPS cells induce macrophage polarization toward an M2-like phenotype which, in turn, increases MLPS cell capability to spread into extracellular matrix and to cross endothelial monolayers. The identification of M2-like polarization factors secreted by MLPS cells may allow to develop novel targeted therapies counteracting MLPS progression.

6.
Eur J Surg Oncol ; 47(10): 2609-2617, 2021 10.
Article in English | MEDLINE | ID: mdl-34083080

ABSTRACT

INTRODUCTION: Bone metastases are frequent in patients with cancer. Electrochemotherapy (ECT) is a minimally invasive treatment. Preclinical and clinical studies supported the use of ECT in patients with metastatic bone disease (MBD). The purposes of this multicentre study are to confirm the safety and efficacy of ECT, and to identify appropriate operating procedures in different MBD conditions. MATERIALS AND METHODS: 102 patients were treated in 11 Centres and recorded in the REINBONE registry (a shared database protected by security passwords): clinical and radiological information, ECT session, adverse events, response, quality of life indicators and duration of follow-up were registered. RESULTS: 105 ECT sessions were performed (one ECT session in 99 patients, two ECT sessions in 3 patients). 24 patients (23.5%) received a programmed intramedullary nail after ECT, during the same surgical procedure. Mean follow-up was 5.9 ± 5.1 months (range 1.5-52). The response to treatment by RECIST criteria was 40.4% objective responses, 50.6% stable disease and 9% progressive disease. According to PERCIST criteria the response was: 31.4% OR; 51.7% SD, 16.9% PD with no significant differences between the 2 criteria. Diagnosis of breast cancer and ECOG values 0-1 were significantly associated to objective response. A significant decrease in pain intensity and significant better quality of life was observed after ECT session at follow-up. CONCLUSION: The results are encouraging on pain and tumour local control. ECT proved to be an effective and safe treatment for MBD and it should be considered as an alternative treatment as well as in combination with radiation therapy.


Subject(s)
Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Electrochemotherapy/methods , Fractures, Spontaneous/surgery , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Bone Nails , Bone Neoplasms/complications , Bone Neoplasms/surgery , Cancer Pain/drug therapy , Cancer Pain/etiology , Disease Progression , Electrochemotherapy/adverse effects , Female , Fracture Fixation, Intramedullary , Fractures, Spontaneous/etiology , Humans , Male , Middle Aged , Prospective Studies , Quality of Life , Response Evaluation Criteria in Solid Tumors
7.
J Transl Med ; 19(1): 173, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33902630

ABSTRACT

Sarcomas are rare, ubiquitous and heterogeneous tumors usually treated with surgery, chemotherapy, target therapy, and radiotherapy. However, 25-50% of patients experience local relapses and/or distant metastases after chemotherapy with an overall survival about 12-18 months. Recently, immuno-therapy has revolutionized the cancer treatments with initial indications for non-small cell lung cancer (NSCLC) and melanoma (immune-checkpoint inhibitors).Here, we provide a narrative review on the topic as well as a critical description of the currently available trials on immunotherapy treatments in patients with sarcoma. Given the promising results obtained with anti-PD-1 monoclonal antibodies (pembrolizumab and nivolumab) and CAR-T cells, we strongly believe that these new immunotherapeutic approaches, along with an innovative characterization of tumor genetics, will provide an exciting opportunity to ameliorate the therapeutic management of sarcomas.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Sarcoma , Humans , Immunotherapy , Neoplasm Recurrence, Local , Sarcoma/therapy
8.
Cells ; 9(4)2020 04 24.
Article in English | MEDLINE | ID: mdl-32344648

ABSTRACT

Chondrosarcomas (CHS) are malignant cartilaginous neoplasms with diverse morphological features, characterized by resistance to chemo- and radiation therapies. In this study, we investigated the role of tumor-associated macrophages (TAM)s in tumor tissues from CHS patients by immunohistochemistry. Three-dimensional organotypic co-cultures were set up in order to evaluate the contribution of primary human CHS cells in driving an M2-like phenotype in monocyte-derived primary macrophages, and the capability of macrophages to promote growth and/or invasiveness of CHS cells. Finally, with an in vivo model of primary CHS cells engrafted in nude mice, we tested the ability of a potent peptide inhibitor of cell migration (Ac-d-Tyr-d-Arg-Aib-d-Arg-NH2, denoted RI-3) to reduce recruitment and infiltration of monocytes into CHS neoplastic lesions. We found a significant correlation between alternatively activated M2 macrophages and intratumor microvessel density in both conventional and dedifferentiated CHS human tissues, suggesting a link between TAM abundance and vascularization in CHS. In 3D and non-contact cu-culture models, soluble factors produced by CHS induced a M2-like phenotype in macrophages that, in turn, increased motility, invasion and matrix spreading of CHS cells. Finally, we present evidence that RI-3 successfully prevent both recruitment and infiltration of monocytes into CHS tissues, in nude mice.


Subject(s)
Chondrosarcoma/pathology , Monocytes/pathology , Tumor-Associated Macrophages/pathology , Adult , Aged , Animals , Antigens, CD/metabolism , Collagen/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunophenotyping , Male , Mice, Nude , Microvessels/pathology , Middle Aged , Monocytes/drug effects , Phenotype , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , THP-1 Cells , Time Factors , Tumor-Associated Macrophages/drug effects
9.
Br J Cancer ; 121(11): 979-982, 2019 11.
Article in English | MEDLINE | ID: mdl-31666667

ABSTRACT

Selection of cancer patients for treatment with immune checkpoint inhibitors remains a challenge due to tumour heterogeneity and variable biomarker detection. PD-L1 expression in 24 surgical chordoma specimen was determined immunohistochemically with antibodies 28-8 and E1L3N. The ability of patient-derived organoids to detect treatment effects of nivolumab was explored by quantitative and qualitative immunofluorescence and FACS analysis. The more sensitive antibody, E1L3N (ROC = 0.896, p = 0.001), was associated with greater tumour diameters (p = 0.014) and detected both tumour cells and infiltrating lymphocytes in 54% of patients, but only 1-15% of their cells. Organoids generated from PD-L1-positive patients contained both tumour cells and PD-1/CD8-positive lymphocytes and responded to nivolumab treatment with marked dose-dependent diameter reductions of up to 50% and increased cell death in both PD-L1-positive and negative organoids. Patient-derived organoids may be valuable to predict individual responses to immunotherapy even in patients with low or no immunohistochemical PD-L1 expression.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Chordoma/metabolism , Drug Discovery/methods , Immunotherapy/methods , Nivolumab/pharmacology , Organoids/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Aged , Aged, 80 and over , Apoptosis/drug effects , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/drug effects , Chordoma/pathology , Chordoma/surgery , Female , Humans , Immunohistochemistry/methods , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Models, Biological , Programmed Cell Death 1 Receptor/metabolism
10.
Acta Biomed ; 90(4): 587-594, 2019 12 23.
Article in English | MEDLINE | ID: mdl-31910191

ABSTRACT

We describe radiographic, contrast-enhanced MDCT and MRI findings with pathologic correlations of an unusual recurrence of tumoral calcinosis, also called Teutschlander disease. The disease was silent in the first decade of life, when it appeared with elbows recurring lesions, until the seventh decade of life, when a left hip active growth lesion developed. A review about tumoral calcinosis pathogenesis, clinical course and imaging differential diagnosis is reported. (www.actabiomedica.it).


Subject(s)
Calcinosis/diagnostic imaging , Hyperostosis, Cortical, Congenital/diagnostic imaging , Hyperphosphatemia/diagnostic imaging , Magnetic Resonance Imaging , Multidetector Computed Tomography , Humans , Male , Middle Aged , Recurrence
11.
Int J Mol Sci ; 19(2)2018 02 21.
Article in English | MEDLINE | ID: mdl-29466296

ABSTRACT

Cancer treatment is rapidly evolving toward personalized medicine, which takes into account the individual molecular and genetic variability of tumors. Sophisticated new in vitro disease models, such as three-dimensional cell cultures, may provide a tool for genetic, epigenetic, biomedical, and pharmacological research, and help determine the most promising individual treatment. Sarcomas, malignant neoplasms originating from mesenchymal cells, may have a multitude of genomic aberrations that give rise to more than 70 different histopathological subtypes. Their low incidence and high level of histopathological heterogeneity have greatly limited progress in their treatment, and trials of clinical sarcoma are less frequent than trials of other carcinomas. The main advantage of 3D cultures from tumor cells or biopsy is that they provide patient-specific models of solid tumors, and they overcome some limitations of traditional 2D monolayer cultures by reflecting cell heterogeneity, native histologic architectures, and cell-extracellular matrix interactions. Recent advances promise that these models can help bridge the gap between preclinical and clinical research by providing a relevant in vitro model of human cancer useful for drug testing and studying metastatic and dormancy mechanisms. However, additional improvements of 3D models are expected in the future, specifically the inclusion of tumor vasculature and the immune system, to enhance their full ability to capture the biological features of native tumors in high-throughput screening. Here, we summarize recent advances and future perspectives of spheroid and organoid in vitro models of rare sarcomas that can be used to investigate individual molecular biology and predict clinical responses. We also highlight how spheroid and organoid culture models could facilitate the personalization of sarcoma treatment, provide specific clinical scenarios, and discuss the relative strengths and limitations of these models.


Subject(s)
Precision Medicine , Sarcoma/pathology , Spheroids, Cellular/pathology , Animals , Humans , Models, Biological , Tumor Cells, Cultured , Tumor Microenvironment
12.
Oncotarget ; 8(49): 85040-85053, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29156702

ABSTRACT

Giant cell tumors of bone (GCTB) are rare sarcomas with a high rate of unpredictable local relapse. Studies suggest that surgical methods affect recurrence, supporting the idea that local disease develops from re-growth of residual cancer cells. To identify early prognostic markers of individual risk of recurrence, we evaluated the effect of post-surgery fluids from a cohort of GCTB patients on growth of primary and established sarcoma cell lines, and mice xenograph. Post-surgery fluids increased cell growth and enhanced expression of CD44++, the principal receptor for the extracellular matrix component hyaluronan and the mesenchymal stem marker CD117+. Cancer cells became highly invasive and tumorigenic, acquiring stemness properties, and activated AKT/mTOR pathway. Prolonged stimulation with post-surgery fluids down-regulated the mesenchymal gene TWIST1 and Vimentin protein, and transdifferentiated cells into tubule-like structures positive to the endothelial markers VE-Cadherin and CD31+. In mice, post-surgery fluids gave rise to larger and more vascularized tumors than control, while in patients AKT/mTOR pathway activation was associated with recurrence by logistic regression (Kaplan-Meier; P<0.001). These findings indicate that post-surgery fluids are an adjuvant in mechanisms of tumor regrowth, increasing stem cell growth and AKT/mTOR activity.

13.
Crit Rev Oncog ; 22(1-2): 99-107, 2017.
Article in English | MEDLINE | ID: mdl-29604939

ABSTRACT

Yin Yang 1 (YY1) belongs to the polycomb group (PcG) of proteins that modify chromatin epigenetically during dynamic regulation of their target genes. The predominant feature of YY1 is the zinc finger, an ancient structural motif that mediates protein-protein interactions and is capable of interacting with both DNA and RNA. Evidence reveals that YY1 acts predominantly as an epigenetic modulator, influencing the activity and/or localization of epigenetic modifiers molecules such as DNA methylation transferases, histone deacetylases, or non-coding RNAs. Deregulation of the epigenome is observed frequently in a variety of cancer types and is often correlated directly with advanced metastatic stages and poor prognosis. In this review, we address the current understanding of YY1 as a recruiter of epi-modifier molecules in the mechanism of aberrant regulation of target genes as a part of the metastatic cascade.


Subject(s)
DNA Methylation/genetics , DNA/genetics , Neoplasms/genetics , YY1 Transcription Factor/genetics , Chromatin/genetics , Epigenomics , Humans , MicroRNAs/genetics , Neoplasm Metastasis , RNA/genetics , Signal Transduction/genetics , Zinc Fingers/genetics
14.
Oncotarget ; 7(34): 54474-54487, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27323409

ABSTRACT

The receptor for the urokinase-type plasminogen activator (uPAR) is a widely recognized master regulator of cell migration and uPAR88-92 is the minimal sequence required to induce cell motility and angiogenesis by interacting with the formyl peptide receptor type 1 (FPR1). In this study, we present evidence that the cyclization of the uPAR88-92 sequence generates a new potent inhibitor of migration, and extracellular matrix invasion of human osteosarcoma and chondrosarcoma cells expressing comparable levels of FPR1 on cell surface. In vitro, the cyclized peptide [SRSRY] prevents formation of capillary-like tubes by endothelial cells co-cultured with chondrosarcoma cells and trans-endothelial migration of osteosarcoma and chondrosarcoma cells. When chondrosarcoma cells were subcutaneously injected in nude mice, tumor size, intra-tumoral microvessel density and circulating tumor cells in blood samples collected before the sacrifice, were significantly reduced in animals treated daily with i.p-administration of 6 mg/Kg [SRSRY] as compared to animals treated with vehicle only. Our findings indicate that [SRSRY] prevents three key events occurring during the metastatic process of osteosarcoma and chondrosarcoma cells: the extracellular matrix invasion, the formation of a capillary network and the entry into bloodstream.


Subject(s)
Bone Neoplasms/blood supply , Chondrosarcoma/blood supply , Neovascularization, Pathologic/drug therapy , Osteosarcoma/blood supply , Peptides, Cyclic/therapeutic use , Receptors, Urokinase Plasminogen Activator/therapeutic use , Animals , Cell Line, Tumor , Cell Movement , Chondrosarcoma/pathology , Female , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Osteosarcoma/pathology , Receptors, Formyl Peptide/physiology
15.
Sarcoma ; 2016: 6862090, 2016.
Article in English | MEDLINE | ID: mdl-27019606

ABSTRACT

Background. In several prospective and retrospective studies, weekly paclitaxel showed promising activity in patients with angiosarcoma. Patients and Methods. Our study was originally designed as a prospective, phase II multicenter trial for patients younger than 75, with ECOG performance status 0-2, affected by locally advanced or metastatic angiosarcoma. Patients received paclitaxel 80 mg/m(2) intravenously, at days 1, 8, and 15 every 4 weeks, until disease progression or unacceptable toxicity. Primary endpoint was objective response. Results. Eight patients were enrolled but, due to very slow accrual, the trial was prematurely stopped and further 10 patients were retrospectively included in the analysis. Out of 17 evaluable patients, 6 patients obtained an objective response (5 partial, 1 complete), with an objective response rate of 35% (95% confidence interval 17%-59%). Of note, five responses were obtained in pretreated patients. In the paper, details of overall survival, progression-free survival, and tolerability are reported. Conclusions. In this small series of patients with locally advanced or metastatic angiosarcoma, weekly paclitaxel was confirmed to be well tolerated and active even in pretreated patients.

16.
In Vivo ; 29(5): 533-40, 2015.
Article in English | MEDLINE | ID: mdl-26359411

ABSTRACT

BACKGROUND/AIM: Giant cell tumors are mostly benign but locally aggressive tumors. The excision of bone tumors can result in large defects, therefore bone reconstruction is still one the most demanding procedures in orthopedic surgery. Our study addresses the opportunity for improving surgical outcome by employing ß-tricalcium phosphate (ß-TCP) with platelet-rich plasma (PRP) at the surgical site. PATIENTS AND METHODS: We included 16 patients with giant cell tumors. After adjuvant therapy, the cavity was reconstructed with ß-TCP, bone graft material (ActifuserR Granules Baxter) and platelet gel application. RESULTS: Our explorative analysis suggests a positive effect of PRP on surgical outcome in patients with giant cell tumors treated with curettage. CONCLUSION: Use of platelet gel as an adjuvant significantly reduces the time required for bone healing following intralesional treatment of benign giant cell tumors, and achieves good functional results without promoting local recurrence.


Subject(s)
Blood Transfusion, Autologous , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Giant Cell Tumor of Bone/pathology , Giant Cell Tumor of Bone/therapy , Plastic Surgery Procedures , Platelet Transfusion , Adolescent , Adult , Aged , Bone Neoplasms/diagnostic imaging , Curettage , Female , Follow-Up Studies , Giant Cell Tumor of Bone/diagnostic imaging , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local , Platelet Transfusion/methods , Tomography, X-Ray Computed , Treatment Outcome , Young Adult
17.
BMC Cancer ; 15: 424, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25998535

ABSTRACT

BACKGROUND: We recently developed a clinical decision support tool, capable of estimating the likelihood of survival at 3 and 12 months following surgery for patients with operable skeletal metastases. After making it publicly available on www.PATHFx.org , we attempted to externally validate it using independent, international data. METHODS: We collected data from patients treated at 13 Italian orthopaedic oncology referral centers between 2010 and 2013, then applied to PATHFx, which generated a probability of survival at three and 12-months for each patient. We assessed accuracy using the area under the receiver-operating characteristic curve (AUC), clinical utility using Decision Curve Analysis (DCA), and compared the Italian patient data to the training set (United States) and first external validation set (Scandinavia). RESULTS: The Italian dataset contained 287 records with at least 12 months follow-up information. The AUCs for the three-month and 12-month estimates was 0.80 and 0.77, respectively. There were missing data, including the surgeon's estimate of survival that was missing in the majority of records. Physiologically, Italian patients were similar to patients in the training and first validation sets. However notable differences were observed in the proportion of those surviving three and 12-months, suggesting differences in referral patterns and perhaps indications for surgery. CONCLUSIONS: PATHFx was successfully validated in an Italian dataset containing missing data. This study demonstrates its broad applicability to European patients, even in centers with differing treatment philosophies from those previously studied.


Subject(s)
Bone Neoplasms/mortality , Bone Neoplasms/secondary , Decision Support Techniques , Models, Statistical , Aged , Bone Neoplasms/diagnosis , Bone Neoplasms/therapy , Female , Humans , Italy , Male , Middle Aged , ROC Curve , Reproducibility of Results , Scandinavian and Nordic Countries , Survival Analysis , United States
18.
Ann Thorac Surg ; 98(1): 291-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24857855

ABSTRACT

BACKGROUND: New materials (NM) such as titanium plates, cryopreserved grafts, and acellular collagen matrices are being increasingly used for chest wall reconstruction as a result of improved incorporation while maintaining structural stability and reduced need for removal from infected areas. Direct comparisons between NM and conventional materials (CM) in terms of local morbidity and need for prosthesis removal are lacking. METHODS: Between January 2005 and July 2013, 109 procedures were performed to remove chest wall tumors in 86 patients. Of these, 32 underwent complex chest wall reconstructions owing to either recurrence, defect extension (greater than 3 ribs or >100 cm2) or local conditions (ie, previous irradiation or infection). New materials and CM (ie, polytetrafluoroethylene and methyl methacrylate) were used in 17 (53%) and 15 (47%) patients, respectively. Of the 32 patients included in the high complexity group, 23 patients did not exhibit any postoperative complications (72%). However, 9 patients (28%) underwent both a first and a second reoperation after a median interval of 4 months from the first procedure (range, 7 days to 60 months). Vacuum-assisted closure (VAC) was instituted in all patients as a means to control sepsis and facilitate space obliteration with healthy tissue. RESULTS: In 7 patients the reason for reintervention was local wound complications. In 4 of 7 patients, the prosthesis had to be removed (3 CM and 1 NM, 4.6% of the whole series; 12.5% in the high complexity group, 5.9% for NM and 20% for CM). The median time to complete chest wall healing after VAC in patients with local sepsis was 14 months (range, 5 to 60 months). All patients are currently alive and well except for 1 who died 11 months after complete chest wall healing as a result of dissemination of metastatic chondrosarcoma. At univariate analysis, predictors of overall and grade 2 or less morbidity according to the Common Terminology Criteria for Adverse Events version 4.0 were first (p=0.038) and second (p=0.015) redo operations. Conversely, patients with a body mass index of less than 25 kg/m2 (p=0.049) undergoing one (p=0.032) or two reconstructions (p=0.00047) with combined materials (p=0.00029) were more likely to experience local wound complications and require VAC. On multiple regression analysis, redo operations (first, p=0.032; second, p=0.00047) and the use of combined (synthetic and biologic) materials (p=0.0029) were confirmed to be related to an increased incidence of wound complications. CONCLUSIONS: Multiple redo operations after complex chest wall reconstruction performed with a combination of NM and CM may be associated with an increased incidence of local wound complications. Nevertheless, in these cases, the use of NM and VAC yielded a low rate (5.8% versus 20% with CM) of prosthesis removal while achieving complete wound healing.


Subject(s)
Negative-Pressure Wound Therapy/statistics & numerical data , Prostheses and Implants , Surgical Wound Infection/epidemiology , Surgical Wound Infection/prevention & control , Thoracic Neoplasms/surgery , Thoracoplasty/adverse effects , Adolescent , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Italy/epidemiology , Male , Middle Aged , Morbidity/trends , Retrospective Studies , Thoracoplasty/instrumentation , Treatment Outcome , Young Adult
19.
Cancers (Basel) ; 5(2): 404-17, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-24216983

ABSTRACT

Cooperation between endothelial cells and bone in bone remodelling is well established. In contrast, bone microvasculature supporting the growth of primary tumors and metastasis is poorly understood. Several antiangiogenic agents have recently been undergoing trials, although an extensive body of clinical data and experimental research have proved that angiogenic pathways differ in each tumor type and stage. Here, for the first time, we characterize at the molecular and functional level tumor endothelial cells from human bone sarcomas at different stages of disease and with different histotypes. We selected a CD31+ subpopulation from biopsies that displayed the capability to grow as adherent cell lines without vascular endothelial growth factor (VEGF). Our findings show the existence in human primary bone sarcomas of highly proliferative endothelial cells expressing CD31, CD44, CD105, CD146 and CD90 markers. These cells are committed to develop capillary-like structures and colony formation units, and to produce nitric oxide. We believe that a better understanding of tumor vasculature could be a valid tool for the design of an efficacious antiangiogenic therapy as adjuvant treatment of sarcomas.

20.
Oncol Rep ; 30(6): 2579-86, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24085196

ABSTRACT

Liposarcoma (LPS) is the most common soft tissue neoplasm in adults and is characterized by neoplastic adipocyte proliferation. Some subtypes of LPSs show aberrations involving the chromosome 12. The most frequent are t(12;16) (q13;p11) present in more than 90% of myxoid LPSs and 12q13-15 amplification in well-differentiated and dedifferentiated LPSs. In this region, there are important oncogenes such as CHOP (DDIT3), GLI, MDM2, CDK4, SAS, HMGA2, but also the HOXC locus, involved in development and tumor progression. In this study, we evaluated the expression of HOXC13, included in this chromosomal region, in a series of adipocytic tumors. We included 18 well-differentiated, 4 dedifferentiated, 11 myxoid and 6 pleomorphic LPSs as well as 13 lipomas in a tissue microarray. We evaluated the HOXC13 protein and gene expression by immunohistochemistry and quantitative PCR. Amplification/translocation of the 12q13-15 region was verified by FISH. Immunohistochemical HOXC13 overexpression was observed in all well-differentiated and dedifferentiated LPSs, all characterized by the chromosome 12q13-15 amplification, and confirmed by quantitative PCR analysis. In conclusion, our data show a deregulation of the HOXC13 marker in well­differentiated and dedifferentiated LPSs, possibly related to 12q13-15 chromosomal amplification.


Subject(s)
Cell Differentiation/genetics , Chromosomes, Human, Pair 12/genetics , Homeodomain Proteins/genetics , Liposarcoma/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Chromosome Aberrations , Female , Gene Amplification , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/biosynthesis , Humans , In Situ Hybridization, Fluorescence , Liposarcoma/pathology , Male , Middle Aged , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...