Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Int J Mol Sci ; 23(22)2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36430780

ABSTRACT

Trabectedin is used for the treatment of advanced soft tissue sarcomas (STSs). In this study, we evaluated if trabectedin could enhance the efficacy of irradiation (IR) by increasing the intrinsic cell radiosensitivity and modulating tumor micro-environment in fibrosarcoma (HS 93.T), leiomyosarcoma (HS5.T), liposarcoma (SW872), and rhabdomyosarcoma (RD) cell lines. A significant reduction in cell surviving fraction (SF) following trabectedin + IR compared to IR alone was observed in liposarcoma and leiomyosarcoma (enhancement ratio at 50%, ER50: 1.45 and 2.35, respectively), whereas an additive effect was shown in rhabdomyosarcoma and fibrosarcoma. Invasive cells' fraction significantly decreased following trabectedin ± IR compared to IR alone. Differences in cell cycle distribution were observed in leiomyosarcoma and rhabdomyosarcoma treated with trabectedin + IR. In all STS lines, trabectedin + IR resulted in a significantly higher number of γ-H2AX (histone H2AX) foci 30 min compared to the control, trabectedin, or IR alone. Expression of ATM, RAD50, Ang-2, VEGF, and PD-L1 was not significantly altered following trabectedin + IR. In conclusion, trabectedin radiosensitizes STS cells by affecting SF (particularly in leiomyosarcoma and liposarcoma), invasiveness, cell cycle distribution, and γ-H2AX foci formation. Conversely, no synergistic effect was observed on DNA damage repair, neoangiogenesis, and immune system.


Subject(s)
Fibrosarcoma , Leiomyosarcoma , Liposarcoma , Radiation-Sensitizing Agents , Rhabdomyosarcoma , Sarcoma , Soft Tissue Neoplasms , Humans , Trabectedin/pharmacology , Trabectedin/therapeutic use , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , Leiomyosarcoma/drug therapy , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Sarcoma/drug therapy , Sarcoma/pathology , Liposarcoma/drug therapy , Tumor Microenvironment
2.
Med Oncol ; 39(8): 119, 2022 Jun 10.
Article in English | MEDLINE | ID: mdl-35687207

ABSTRACT

Circulating tumor cells detection and ARV7 expression are associated with worse clinical outcomes in metastatic Castration-Resistant Prostate Cancer (mCRPC) undergoing Androgen Receptor Targeted Agents. ARFL, PSMA and PSA may help to refine prognostic models. In our institution, a prospective observational trial testing CTC detection in mCPRC undergoing I line ARTA therapy terminated the planned enrollment in 2020. Here, we present a pre-planned interim analysis with 18 months of median follow-up. RT-qPCR was used to determine the CTC expression of PSA, PSMA, AR and ARV7 before starting ARTA. PSA-drop, Progression-Free and Overall Survival (PFS and OS) and their correlation with CTC detection were reported. Forty-four patients were included. CTC were detected in 43.2% of patients, of whom 8.94% expressed PSA, 15.78% showed ARV7, 63.15% and 73.68% displayed ARFL and PSMA, respectively. Biochemical response was significantly improved in CTC + vs CTC- patients, with median PSA-drop of 18.5 vs 2.5 ng/ml (p = 0.03). After a median follow-up of 18 months, 50% of patients progressed. PFS was significantly longer in CTC- patients (NR vs 16 months). Eight (18.2%) patients died, a non-significant trend in terms of OS was detected in favor of CTC- patients (NR vs 29 months, p = 0.05). AR, PSA and PSMA expression in CTC + had no significant impact on PSA-drop, PFS or OS. PRIMERA-trial confirmed the CTC detection predictive importance in mCRPC patients.


Subject(s)
Antineoplastic Agents , Neoplastic Cells, Circulating , Prostatic Neoplasms, Castration-Resistant , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Humans , Male , Neoplastic Cells, Circulating/pathology , Prospective Studies , Prostate-Specific Antigen , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Treatment Outcome
3.
J Pers Med ; 11(11)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34834449

ABSTRACT

Adrenocortical carcinoma (ACC) is a rare malignancy with poor prognosis when metastatic and scarce treatment options in the advanced stages. In solid tumors, the chemokine CXCL12/CXCR4 axis is involved in the metastatic process. We demonstrated that the human adrenocortex expressed CXCL12 and its cognate receptors CXCR4 and CXCR7, not only in physiological conditions, but also in ACC, where the receptors' expression was higher and the CXCL12 expression was lower than in the physiological conditions. In a small pilot cohort of 22 ACC patients, CXCL12 negatively correlated with tumor size, stage, Weiss score, necrosis, and mitotic activity. In a Kaplan-Meier analysis, the CXCL12 tumor expression significantly predicted disease-free, progression-free, and overall survival. In vitro treatment of the primary ACC H295R and of the metastatic MUC-1 cell line with the PPARγ-ligand rosiglitazone (RGZ) dose-dependently reduced proliferation, resulting in a significant increase in CXCL12 and a decrease in its receptors in the H295R cells only, with no effect on the MUC-1 levels. In ACC mouse xenografts, tumor growth was inhibited by the RGZ treatment before tumor development (prevention-setting) and once the tumor had grown (therapeutic-setting), similarly to mitotane (MTT). This inhibition was associated with a significant suppression of the tumor CXCR4/CXCR7 and the stimulation of human CXCL12 expression. Tumor growth correlated inversely with CXCL12 and positively with CXCR4 expression, suggesting that local CXCL12 may impair the primary tumor cell response to the ligand gradient that may contribute to driving the tumor progression. These findings indicate that CXCL12/CXCR4 may constitute a potential target for anti-cancer agents such as rosiglitazone in the treatment of ACC.

4.
Clin Transl Radiat Oncol ; 23: 60-64, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32435703

ABSTRACT

AIM: In a pilot prospective study, we aimed to test the feasibility and report on the preliminary results on the expression of molecular biomarkers in wound drainage fluids (WDFs) of operated head and neck squamous cell carcinoma (HNSCC) patients. MATERIAL AND METHODS: Nineteen patients undergoing primary tumor resection with en-block neck dissection were enrolled. In postoperative days 1-3, the expression of several biomarkers in WDFs was measured using enzyme-linked immunosorbent assay (ELISA) kits and correlated with clinical and histopathologic features. RESULTS: The expression of stromal cell-derived factor 1 (CXCL-12) was significantly increased in WDFs in presence of lymph node metastases, extranodal extension (ENE), and in case of close resection margins. In addition, Osteopontin expression was significantly increased in presence of ENE, whereas transforming growth factor beta (TGF-ß) detection was significantly reduced. At multivariate analysis, CXCL-2 levels in both day 1 and 3 post-surgery were the only factor which retained significance in the prediction of close surgical margins (p = 0.028 and 0.025 for day 1 and day 3, respectively). Both CXCL-2 and Ostepontin assays were significantly correlated with ENE (p = 0.018 and 0.035 for day 1; 0.052 and 0.025 for day 3, respectively) whereas TGF- ß expression was significant at day 1 only (p = 0.038). CONCLUSIONS: Our pilot study showed that WDFs could qualify as a potential source of relevant postoperative information. Further studies are needed to confirm the prognostic impact of CXCL-12, Osteopontin and TGF-ß expressed in WDFs on the personalized management of HNSCC.

5.
Radiol Med ; 124(4): 282-289, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29582320

ABSTRACT

BACKGROUND: Poly(ADP-ribose) polymerases (PARP) are a large family of enzymes involved in several cellular processes, including DNA single-strand break repair via the base-excision repair pathway. PARP inhibitors exert antitumor activity by both catalytic PARP inhibition and PARP-DNA trapping, moreover PARP inhibition represents a potential synthetic lethal approach against cancers with specific DNA-repair defects. Soft tissue sarcoma (STSs) are a heterogeneous group of mesenchymal tumors with locally destructive growth, high risk of recurrence and distant metastasis. OBJECTIVES: The purpuse of this review is to provide an overview of the main preclinical and clinical data on use of PARPi in STSs and of effect and safety of combination of PARPi with irradiation. RESULTS: Due to numerous genomic alterations in STSs, the DNA damage response pathway can offer an interesting target for biologic therapy. Preclinical and clinical studies showed promising results, with the most robust evidences of PARPi efficacy obtained on Ewing sarcoma bearing EWS-FLI1 or EWS-ERG genomic fusions. The activity of PARP inhibitors resulted potentiated by chemotherapy and radiation. Although mechanisms of synergisms are not completely known, combination of radiation therapy and PARP inhibitors exerts antitumor effect by accumulation of unrepaired DNA damage, arrest in G2/M, activity both on oxic and hypoxic cells, reoxygenation by effect on vessels and promotion of senescence. Early trials have shown a good tolerance profile. CONCLUSIONS: The use of PARP inhibitors in advanced stage STSs, alone or combined in multimodal treatments, is of great interest and warrants further investigations.


Subject(s)
Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Sarcoma/drug therapy , Sarcoma/radiotherapy , Animals , Combined Modality Therapy , DNA Damage , Humans
6.
Radiat Res ; 190(5): 464-472, 2018 11.
Article in English | MEDLINE | ID: mdl-30067444

ABSTRACT

Soft tissue sarcomas (STS) are aggressive tumors with a poor prognosis. Poly(ADP-ribose) polymerase (PARP)-1 inhibitors (PARPi) enhance the cytotoxic effects of radiation. In this study, we evaluated the effect of PARPi on survival and DNA damage of irradiated STS cells. For clonogenic assays, STS cell lines were irradiated with or without olaparib, iniparib or veliparib pretreatment. The effect of PARP inhibition on γ-H2AX and Rad51 foci formation, on PARP-1, phospho-ERK and cleaved caspase-3 protein expression and on cell cycle progression was evaluated on irradiated rhabdomyosarcoma cells pretreated with olaparib. The results from this work showed that PARPi induced significant radiosensitization in STS cells. Rhabdomyosarcoma cells showed the highest increase in radiosensitivity, with a radiosensitization enhancement ratio at 50% survival (ER50) of 3.41 with veliparib. All PARPi exerted a synergistic effect when combined with radiation. Fibrosarcoma cells showed an ER50 of 2.29 with olaparib. Leiomyosarcoma and liposarcoma cells showed their highest ER50 with veliparib (1.71 and 1.84, respectively). In rhabdomyosarcoma, olaparib enhanced the formation of radiation-induced γ-H2AX/Rad51 foci and PARP-1 cleavage, induced slightly increased expression of cleaved caspase-3 and reduced phospho-ERK expression. Moreover, the combination of olaparib and radiation resulted in a significantly enhanced cell cycle arrest in the G2/M phase compared to the two treatments alone. In conclusion, we have shown that PARPi are potent radiosensitizers of human STS cells. These results support the pursuit of further investigations into the effects of PARPi combined with radiation on STS.


Subject(s)
Antineoplastic Agents/therapeutic use , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/therapeutic use , Sarcoma/pathology , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Combined Modality Therapy , DNA Damage/drug effects , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Radiation-Sensitizing Agents/pharmacology , Sarcoma/drug therapy , Sarcoma/radiotherapy , Xenograft Model Antitumor Assays
7.
Clin Transl Radiat Oncol ; 9: 42-47, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29594250

ABSTRACT

INTRODUCTION AND BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) has been increasingly recognized as an immune suppressive malignancy. The efficacy of immune checkpoint inhibitors (ICI's) in the context of recurrent/metastatic (R/M) setting anticipates the possible integration of immunotherapy into the therapeutic armamentarium of locally advanced disease. Durvalumab (DUR) is a humanized monoclonal IgG1, anti-PD-L1 antibody with promising data in R/M HNSCC. The aim of our study is to test the antitumor activity of a combined regimen incorporating an immune checkpoint inhibitor into a conventional bio-radiation strategy for the cure of unfavorable locally advanced HNSCC. METHODS/DESIGN: In this open label, multi-center, single-arm, phase I/II study, enrolled patients will receive Radiotherapy (RT) (69.9 Gy/2.12 Gy in 33 fractions) with concurrent Cetuximab (CTX) (400 mg/m2 1 week before RT start followed by 250 mg/m2 weekly) and DUR (fixed dose of 1500 mg every 4 weeks starting from RT-CTX week 1) followed by adjuvant DUR (to a maximum of 6 months after completion of RT-CTX). Primary endpoint of the study is 2-year progression-free survival (PFS). A safety run-in is planned after the enrollment of first 12, 24 and 36 patients. Patients affected by high-risk (≥N2a or ≥T3, any N) larynx, hypopharynx and HPV negative oropharynx or HPV-positive oropharynx (≥T2, ≥N2b, ≥10 pack/years) will be eligible. DISCUSSION: Conventional intensification strategies failed to provide any benefit for the cure of locally advanced HNSCC. For the still prevalent HPV-negative population and the high risk-HPV positive disease, there is an unmet need for alternative treatment paradigms. Potentially, the inhibition of the PD-1/PD-L1 checkpoint may synergize with both CTX and RT through immunologic interplay, ultimately aiming to reverse the HNSCC-induced immune suppression. The DUCRO study will seek to demonstrate if such a strategy may be safe and active. TRIAL REGISTRATION: NCT number: NCT03051906Eudract number: 2016-004668-20.

8.
Head Neck ; 40(3): 577-583, 2018 03.
Article in English | MEDLINE | ID: mdl-29155481

ABSTRACT

BACKGROUND: Oral mucositis is a side effect of treatment regimens containing 5-fluorouracil (5-FU). The purpose of this study was to present our evaluation to see if rosiglitazone (RGZ) protected normal tissues from chemotherapy-induced oral mucositis. METHODS: C57BL/6J mice were treated with 5-FU for 5 days, with or without RGZ. Mice were euthanized after 5, 8, 11, or 15 days, and mucosal segments were collected. RESULTS: The RGZ did not affect the 5-FU-induced decrease in mouse body weight. The 5-FU caused loss of epithelial architecture, collagen fiber impairment, and inflammatory infiltration. The RGZ reduced leukocyte infiltration, preserved tissue structure, and dampened the 5-FU-induced expression of p53 and matrix metalloproteinase (Mmp)-2 after 5 days, and of Mmp-2 and interleukin (Il-1ß after 15 days. The RGZ inhibited the 5-FU-induced increase of transforming growth factor-beta (TGF-ß) and nuclear factor-kappa B (NF-κB) proteins and restored collagen protein levels. CONCLUSION: The RGZ had a protective effect on oral mucosa damaged by chemotherapy. These data encourage the further study of RGZ for the prevention of 5-FU-induced mucositis in patients with cancer.


Subject(s)
Fluorouracil/adverse effects , PPAR gamma/agonists , Rosiglitazone/pharmacology , Stomatitis/chemically induced , Animals , Blotting, Western , Cytokines/metabolism , Fluorouracil/pharmacology , Mice , Mice, Inbred C57BL , Mouth Mucosa/drug effects , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Real-Time Polymerase Chain Reaction , Stomatitis/prevention & control
9.
United European Gastroenterol J ; 5(2): 218-226, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28344789

ABSTRACT

OBJECTIVE: Because of its anti-inflammatory, anti-fibrotic, anti-apoptotic and anti-neoplastic properties, the PPAR-γ agonist rosiglitazone is an interesting drug for investigating for use in the prevention and treatment of radiation-induced intestinal damage. We aimed to evaluate the radioprotective effect of rosiglitazone in a murine model of acute intestinal damage, assessing whether radioprotection is selective for normal tissues or also occurs in tumour cells. METHODS: Mice were total-body irradiated (12 Gy), with or without rosiglitazone (5 mg/kg/day). After 24 and 72 hours, mice were sacrificed and the jejunum was collected. HT-29 human colon cancer cells were irradiated with a single dose of 2 (1000 cells), 4 (1500 cells) or 6 (2000 cells) Gy, with or without adding rosiglitazone (20 µM) 1 hour before irradiation. HT-29-xenografted CD1 mice were irradiated (16 Gy) with or without rosiglitazone; tumour volumes were measured for 33 days. RESULTS: Rosiglitazone markedly reduced histological signs of altered bowel structures, that is, villi shortening, submucosal thickening, necrotic changes in crypts, oedema, apoptosis, and inflammatory infiltrate induced by irradiation. Rosiglitazone significantly decreased p-NF-kB p65 phosphorylation and TGFß protein expression at 24 and 72 hours post-irradiation and significantly decreased gene expression of Collagen1, Mmp13, Tnfα and Bax at 24 hours and p53 at 72 hours post-irradiation. Rosiglitazone reduced HT-29 clonogenic survival, but only produced a slight reduction of xenograft tumour growth. CONCLUSION: Rosiglitazone exerts a protective effect on normal tissues and reduces alterations in bowel structures and inflammation in a radiation-induced bowel toxicity model, without interfering with the radiation effect on HT-29 cancer cells. PPAR-γ agonists should be further investigated for their application in abdominal and pelvic irradiation.

10.
Crit Rev Oncol Hematol ; 112: 1-10, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28325250

ABSTRACT

Major advances in the knowledge of cancer biology and its interactions with tumor immune environment led to the emergence, in the last five years of new immunotherapy-based treatment strategies in cancer patients. At the same time, improvement in radiation technique and progress in radiobiology allowed in the last decade to expand the applications of radiotherapy in a growing number of settings. At present, there are strong theoretical basis to propose immune-enhanced radiation therapy that may represent in the future a new paradigm of treatment, combining the intrinsic power of radiotherapy to elicit a specific, systemic, tumor-directed immune response with modern highly conformal and precise dose delivery, in order to maximize response at the major site of disease and obtain durable disease control. The aim of this review is to describe the principal mechanisms of immune modulation of response to radiation and investigational strategies to harness the potential of radiation-inducible immune response: radiation therapy is expected to be not just a local treatment but the cornerstone of a multimodal strategy that might achieve long-lasting tumor remission at the primary site and systemic efficacy metastatic lesions.


Subject(s)
Neoplasms/immunology , Neoplasms/radiotherapy , Radiotherapy/methods , Radiotherapy/trends , Humans , Immunotherapy/methods
11.
Oral Oncol ; 64: 52-58, 2017 01.
Article in English | MEDLINE | ID: mdl-28024724

ABSTRACT

BACKGROUND: Due to its anti-inflammatory, antifibrotic and antineoplastic properties, the PPAR gamma agonist rosiglitazone is of interest in prevention and therapy of radiation-induced toxicities. We aimed to evaluate the radioprotective effect of rosiglitazone in a mouse model of radiation-induced oral mucositis. MATERIAL AND METHODS: Oral mucositis was obtained by irradiation of the oral region of C57BL/6J mice, pretreated or not with rosiglitazone. Mucositis was assessed by macroscopic scoring, histology and molecular analysis. Tumor xenograft was obtained by s.c. injection of Hep-2 cells in CD1 mice. Tumor volume was measured twice a week to evaluate effect of rosiglitazone alone and combined with radiotherapy. RESULTS: Irradiated mice showed typical features of oral mucositis, such as oedema and reddening, reaching the peak of damage after 12-15days. Rosiglitazone markedly reduced visible signs of mucositis and significantly reduced the peak. Histological analysis showed the presence of an inflammatory cell infiltrate after irradiation; the association with rosiglitazone noticeably reduced infiltration. Rosiglitazone significantly inhibited radiation-induced tnfα, Il-6 and Il-1ß gene expression. Rosiglitazone controlled the increase of TGF-ß and NF-kB p65 subunit proteins induced by irradiation, and enhanced the expression of catalase. Irradiation and rosiglitazone significantly reduced tumor volume as compared to control. Rosiglitazone did not protect tumor from the therapeutic effect of radiation. CONCLUSION: Rosiglitazone exerted a protective action on normal tissues in radiation-induced mucositis. Moreover, it showed antineoplastic properties on head-neck carcinoma xenograft model and selective protection of normal tissues. Thus, PPAR gamma agonists should be further investigated as radioprotective agents in head and neck cancer.


Subject(s)
Disease Models, Animal , PPAR gamma/agonists , Radiotherapy/adverse effects , Stomatitis/prevention & control , Animals , Mice , Mice, Inbred C57BL , Stomatitis/etiology
12.
Anticancer Res ; 35(7): 3875-84, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26124333

ABSTRACT

BACKGROUND/AIM: Patients with prostate cancer treated with neoadjuvant androgen ablation experience less radiation-induced intestinal toxicity, mostly due to a reduction of the volume of normal tissue exposed to high radiation doses. We aimed to evaluate if the anti-androgenic drug leuprorelin itself exerts a protective effect on irradiated bowel. MATERIALS AND METHODS: Female, intact and castrated male C57BL/6J mice underwent 12-Gy total body irradiation, with or without a three-month leuprorelin (0.054 mg/kg/month i.p.) pre-treatment. After 24-72 h, mice were sacrificed and intestinal segments collected for histological, immunohistochemical and molecular analyses. RESULTS: Leuprorelin markedly reduced radiation-induced jejunal and colonic histological alterations in mice, increased the number of regenerating crypts vs. irradiation, and reduced radiation-induced nitrotyrosine immunoreactivity. Leuprorelin significantly reduced radiation-induced matrix metallo-proteinase-2 (Mmp2) and -13, collagen 1 and -3, transforming growth factor-beta (Tgfb), p53, interleukin 6 (Il6), and B-cell lymphoma 2 (Bcl2)-associated X protein (Bax) gene expressions, and nuclear factor-kappa B (NFκB) and TGFß protein expression, and hampered radiation-induced BCL2 protein down-regulation. CONCLUSION: Leuprorelin protects mice from radiation-induced intestinal injury, likely through a reduction of tissue oxidative stress. These findings give a biological interpretation to clinical observations of improved intestinal tolerance in patients undergoing androgen ablation before RT.


Subject(s)
Intestines/drug effects , Leuprolide/pharmacology , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/pharmacology , Animals , Down-Regulation/drug effects , Female , Gene Expression Regulation/drug effects , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Radiation Injuries, Experimental/metabolism
13.
Lung Cancer ; 90(3): 405-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26791799

ABSTRACT

PURPOSE/OBJECTIVE(S): Due to its anti-inflammatory, antifibrotic and antineoplastic properties, the PPAR-γ agonist rosiglitazone is of interest in the prevention and therapy of radiation-induced pulmonary injury. We evaluated the radioprotective effects of rosiglitazone in a murine model of pulmonary damage to determine whether radioprotection was selective for normal and tumor tissues. METHODS: Lungs in C57BL/6J mice were irradiated (19 Gy) with or without rosiglitazone (RGZ, 5mg/kg/day for 16 weeks, oral gavage). Computed tomography (CT) was performed and Hounsfield Units (HU) were determined during the observation period. Histological analysis and evaluation of fibrosis/inflammatory markers by western blot were performed at 16 weeks. A549 tumor-bearing CD1 mice were irradiated (16 Gy) with or without RGZ, and tumor volumes were measured at 35 days. RESULTS: Rosiglitazone reduced radiologic and histologic signs of fibrosis, inflammatory infiltrate, alterations to alveolar structures, and HU lung density that was increased due to irradiation. RGZ treatment also significantly decreased Col1, NF-kB and TGF-ß expression and increased Bcl-2 protein expression compared to the irradiation group and reduced A549 clonogenic survival and xenograft tumor growth. CONCLUSIONS: Rosiglitazone exerted a protective effect on normal tissues in radiation-induced pulmonary injury, while irradiated lung cancer cells were not protected in vivo and in vitro. Thus, rosiglitazone could be proposed as a radioprotective agent in the treatment of lung cancer.


Subject(s)
Lung Injury/metabolism , Lung Injury/pathology , PPAR gamma/agonists , Radiation Injuries, Experimental , Animals , Cell Line, Tumor , Cell Survival/radiation effects , Disease Models, Animal , Humans , Lung Injury/diagnostic imaging , Lung Injury/drug therapy , Mice , Radiation, Ionizing , Radiation-Protective Agents/pharmacology , Radiotherapy/adverse effects , Rosiglitazone , Thiazolidinediones/pharmacology , Tomography, X-Ray Computed , Xenograft Model Antitumor Assays
14.
PLoS One ; 8(10): e77745, 2013.
Article in English | MEDLINE | ID: mdl-24204948

ABSTRACT

OBJECTIVE: This study aims to investigate in vitro the effect of the VDR agonist BXL-01-0029 onto IFNγ/TNFα-induced CXCL10 secretion by human skeletal muscle cells compared to elocalcitol (VDR agonist), methylprednisolone, methotrexate, cyclosporin A, infliximab and leflunomide; to assess in vivo circulating CXCL10 level in subjects at time of diagnosis with IMs, before therapy, together with TNFα, IFNγ, IL-8, IL-6, MCP-1, MIP-1ß and IL-10, vs. healthy subjects. METHODS: Human fetal skeletal muscle cells were used for in vitro studies; ELISA and Bio-Plex were used to measure cell supernatant and IC50 determination or serum cytokines; Western blot and Bio-Plex were for cell signaling analysis. RESULTS: BXL-01-0029 decreased with the highest potency IFNγ/TNFα-induced CXCL10 protein secretion and targeted cell signaling downstream of TNFα in human skeletal muscle cells; CXCL10 level was the highest in sera of subjects diagnosed with IMs before therapy and the only one significantly different vs. healthy controls. CONCLUSIONS: Our in vitro and in vivo data, while confirm the relevance of CXCL10 in IMs, suggested BXL-01-0029 as a novel pharmacological tool for IM treatment, hypothetically to be used in combination with the current immunosuppressants to minimize side effects.


Subject(s)
Myositis/drug therapy , Receptors, Calcitriol/agonists , Adult , Aged , Aged, 80 and over , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CCL4/metabolism , Chemokine CXCL10/metabolism , Female , Humans , Immunosuppressive Agents/pharmacology , Interferon-gamma/metabolism , Interleukins/metabolism , Male , Middle Aged , Muscle Cells/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myositis/metabolism , Receptors, Calcitriol/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
15.
Eur J Cell Biol ; 91(2): 139-49, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22176919

ABSTRACT

Inflammatory myopathies (IMs) are systemic diseases characterized by a T helper (Th) 1 type inflammatory response and cell infiltrates within skeletal muscles. The mainstay of treatment is drugs aimed at suppressing the immune system - corticosteroids and immunosuppressants. About 25% of patients are non-responders. Skeletal muscle cells seem actively involved in the immune-inflammatory response and not only a target; understanding the molecular bases of IMs might help drug development strategies. Within muscles the interaction between the chemokine interferon (IFN)γ inducible 10 kDa protein, CXCL10 or IP-10, and its specific receptor CXCR3, present on Th1 type infiltrating cells, likely plays a pivotal role, potentially offering the opportunity for therapeutic intervention. We aimed to clarify the involvement of human skeletal muscle cells in inflammatory processes in terms of CXCL10 secretion, to elucidate the engaged molecular mechanism(s) and, finally, to evaluate muscular cell responses, if any, to some immunosuppressants routinely used in IM treatment, such as methylprednisolone, methotrexate, cyclosporin A and Infliximab. We first isolated and characterized human fetal skeletal muscle cells (Hfsmc), which expressed the specific lineage markers and showed the competence to react in the context of an in vitro alloresponse. CXCL10 protein secretion by Hfsmc was similarly induced by the inflammatory cytokines interferon (IFN)γ and tumor necrosis factor (TNF)α, above undetectable control levels, through the activation of Stat1 and NF-kB pathways, respectively; CXCL10 secretion was significantly magnified by cytokine combination, and this synergy was associated to a significant up-regulation of TNFαRII; cytokine-induced CXCL10 secretion was considerably affected only by Infliximab. Our data suggested that human skeletal muscle cells might actively self-promote muscular inflammation by eliciting CXCL10 secretion, which is known to amplify Th1 cell tissue infiltration in vivo. In conclusion, we sustain that pharmacological targeting of CXCL10 within muscular cells might contribute to keep in control pro-Th1 polarization of the immune/inflammatory response.


Subject(s)
Chemokine CXCL10/metabolism , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Myositis/metabolism , Receptors, CXCR3/metabolism , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Chemokine CXCL10/immunology , Cyclosporine/pharmacology , Fetus , Humans , Immunosuppressive Agents/pharmacology , Infliximab , Interferon-gamma/pharmacology , Lymphocyte Activation , Methylprednisolone/pharmacology , Muscle Cells/drug effects , Muscle Cells/immunology , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Myositis/immunology , NF-kappa B/metabolism , Receptors, CXCR3/immunology , STAT1 Transcription Factor/metabolism , Signal Transduction , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/pharmacology
16.
Transpl Int ; 23(9): 914-23, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20302596

ABSTRACT

During kidney allograft rejection, CXC chemokine ligand 10 (CXCL10)-CXC chemokine receptor 3 (CXCR3) trafficking between peripheral blood and tissues initiates alloresponse and perpetuates a self-inflammatory loop; thus, CXCL10-CXCR3 axis could represent a pharmacologic target. In this perspective, immunosuppressors targeting graft-resident cells, beside immune cells, could be very advantageous. Vitamin D receptor (VDR) agonists exhibit considerable immunomodulatory properties. This study aimed to investigate whether elocalcitol and BXL-01-0029 could decrease the expression of CXCL10 in activated renal tubular cells in vitro and thus be useful in kidney allograft rejection treatment. Experiments were performed in human tubular renal cells stimulated with interferon-gamma + tumor necrosis factor-alpha with and without VDR agonists, tacrolimus, sirolimus, hydrocortisone, methylprednisolone, cyclosporin A and mycophenolate mofetil. CXCL10 protein secretion and gene expression were measured by ELISA and by quantitative PCR. Specific inhibitors were used to investigate intracellular pathways involved in tubular cells activation. For IC(50) determination and comparison, dose-response curves with VDR agonists, tacrolimus and mycophenolic acid were performed. Elocalcitol and BXL-01-0029 inhibited CXCL10 secretion by renal cells, without affecting cell viability, while almost all the immunosuppressors were found to be ineffective, except for tacrolimus and mycophenolate mofetil. BXL-01-0029 was the most potent drug and, notably, it was found to be capable of allowing reduction in tacrolimus-inhibitory doses. Our data suggest that BXL-01-0029 could potentially be a dose-reducing agent for conventional immunosuppressors in kidney rejection management.


Subject(s)
Calcitriol/analogs & derivatives , Chemokine CXCL10/biosynthesis , Gene Expression Regulation/drug effects , Immunosuppressive Agents/pharmacology , Kidney Tubules/metabolism , RNA/genetics , Receptors, Calcitriol/agonists , Adult , Calcitriol/pharmacology , Cells, Cultured , Chemokine CXCL10/drug effects , Chemokine CXCL10/genetics , Enzyme-Linked Immunosorbent Assay , Graft Rejection/metabolism , Graft Rejection/prevention & control , Humans , Kidney Transplantation , Kidney Tubules/cytology , Kidney Tubules/drug effects , Polymerase Chain Reaction , Reference Values
17.
Transplantation ; 87(2): 249-55, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19155980

ABSTRACT

BACKGROUND: The detection of acute rejection in heart transplantation remains an important feature of transplant management, especially in the early phase. Frequent surveillance with endomyocardial biopsy is necessary, even though it is an invasive procedure and carries a certain risk. Hence, noninvasive biomarkers able to predict acute rejection could be a further helpful tool in patient management. The interferon-gamma-inducible chemokine CXCL10 is required for initiation and development of graft failure caused by acute or chronic rejection. It has been reported that CXCL10 serum level is predictive of graft loss in kidney graft recipients. In the present study, we investigated whether pretransplant CXCL10 serum level may be a predictive noninvasive biomarker in heart transplant (HTx) recipients, as well. METHODS: Sera from 143 patients undergoing orthotopic heart transplantation were collected before surgery and tested for CXCL10 and CCL22 and compared with serum samples from healthy subjects. RESULTS: We found that basal CXCL10 serum levels in HTx recipients were significantly higher than in healthy subjects, whereas no difference was seen in CCL22 levels. Among HTx recipients, CXCL10 serum levels of rejectors were significantly higher than in nonrejectors. Our results showed that CXCL10 was a significant independent risk factor of several variables and had the highest predictive value for early acute heart rejection, with 160 pg/mL cutoff value. CONCLUSIONS: In HTx recipients, measurement of pretransplant CXCL10 serum levels could be a clinically useful tool for predicting cardiac acute rejection, especially in the early posttransplant period.


Subject(s)
Cardiomyopathies/surgery , Chemokine CXCL10/blood , Graft Rejection/diagnosis , Heart Transplantation/immunology , Acute Disease , Biomarkers/blood , Cardiomyopathies/immunology , Chemokine CCL22/blood , Female , Graft Rejection/immunology , Humans , Male , Middle Aged , Predictive Value of Tests , Preoperative Care , ROC Curve , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors , Up-Regulation
18.
Exp Cell Res ; 314(6): 1337-50, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18291365

ABSTRACT

CXCL10-CXCR3 axis plays a pivotal role in cardiac allograft rejection, so that targeting CXCL10 without inducing generalized immunosuppression may be of therapeutic significance in allotransplantation. Since the role of resident cells in cardiac rejection is still unclear, we aimed to establish reliable human cardiomyocyte cultures to investigate Th1 cytokine-mediated response in allograft rejection. We used human fetal cardiomyocytes (Hfcm) isolated from fetal hearts, obtained after legal abortions. Hfcm expressed specific cardiac lineage markers, specific cardiac structural proteins, typical cardiac currents and generated ventricular action potentials. Thus, Hfcm represent a reliable in vitro tool for allograft rejection research, since they resemble the features of mature cells. Hfcm secreted CXCL10 in response to IFNgamma and TNFalphaalpha; this effect was magnified by cytokine combination. Cytokine synergy was associated to a significant TNFalpha-induced up-regulation of IFNgammaR. The response of Hfcm to some currently used immunosuppressive drugs compared to rosiglitazone, a peroxisome proliferator-activated receptor gamma agonist and Th1-mediated response inhibitor, was also evaluated. Only micophenolic acid and rosiglitazone halved CXCL10 secretion by Hfcm. Given the pivotal role of IFNgamma-induced chemokines in Th1-mediated allograft rejection, these preliminary results suggest that the combined effects of immunosuppressive agents and rosiglitazone could be potentially beneficial to patients receiving heart transplants.


Subject(s)
Graft Rejection/immunology , Heart Transplantation/immunology , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Models, Biological , Action Potentials/drug effects , Biomarkers/metabolism , Cell Lineage/drug effects , Cell Separation , Cell Shape/drug effects , Cells, Cultured , Cytokines/metabolism , Electric Capacitance , Gap Junctions/drug effects , Gap Junctions/metabolism , Humans , Inflammation Mediators/metabolism , Interferon-gamma/pharmacology , Ion Channel Gating/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...