Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Med Chem ; 27(2): 187-215, 2020.
Article in English | MEDLINE | ID: mdl-29956610

ABSTRACT

Chemoprevention is based on the use of non-toxic, pharmacologically active agents to prevent tumor progression. In this regard, natural dietary agents have been described by the most recent literature as promising tools for controlling onset and progression of malignancies. Extensive research has been so far performed to shed light on the effects of natural products on tumor growth and survival, disclosing the most relevant signal transduction pathways targeted by such compounds. Overall, anti-inflammatory, anti-oxidant and cytotoxic effects of dietary agents on tumor cells are supported either by results from epidemiological or animal studies and even by clinical trials. Multiple myeloma is a hematologic malignancy characterized by abnormal proliferation of bone marrow plasma cells and subsequent hypercalcemia, renal dysfunction, anemia, or bone disease, which remains incurable despite novel emerging therapeutic strategies. Notably, increasing evidence supports the capability of dietary natural compounds to antagonize multiple myeloma growth in preclinical models of the disease, underscoring their potential as candidate anti-cancer agents. In this review, we aim at summarizing findings on the anti-tumor activity of dietary natural products, focusing on their molecular mechanisms, which include inhibition of oncogenic signal transduction pathways and/or epigenetic modulating effects, along with their potential clinical applications against multiple myeloma and its related bone disease.


Subject(s)
Multiple Myeloma , Animals , Antineoplastic Agents , Bone Diseases , Bone and Bones , Multiple Myeloma/prevention & control , Multiple Myeloma/therapy , Signal Transduction
2.
Oncotarget ; 7(13): 15868-84, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26908461

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive CNS tumor and is characterized by a very high frequency of clinical relapse after therapy and thus by a dismal prognosis, which strongly compromises patients survival. We have recently identified the small molecule SI113, as a potent and selective inhibitor of SGK1, a serine/threonine protein kinase, that modulates several oncogenic signaling cascades. The SI113-dependent SGK1 inhibition induces cell death, blocks proliferation and perturbs cell cycle progression by modulating SGK1-related substrates. SI113 is also able to strongly and consistently block, in vitro and in vivo, growth and survival of human hepatocellular-carcinomas, either used as a single agent or in combination with ionizing radiations. In the present paper we aim to study the effect of SI113 on human GBM cell lines with variable p53 expression. Cell viability, cell death, caspase activation and cell cycle progression were then analyzed by FACS and WB-based assays, after exposure to SI113, with or without oxidative stress and ionizing radiations. Moreover, autophagy and related reticulum stress response were evaluated. We show here, that i) SGK1 is over-expressed in highly malignant gliomas and that the treatment with SI113 leads to ii) significant increase in caspase-mediated apoptotic cell death in GBM cell lines but not in normal fibroblasts; iii)enhancement of the effects of ionizing radiations; iv) modulation of the response to oxidative reticulum stress; v) induction of cytotoxic autophagy. Evidence reported here underlines the therapeutic potential of SI113 in GBM, suggesting a new therapeutic strategy either alone or in combination with radiotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Glioblastoma/drug therapy , Oxidative Stress/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemoradiotherapy/methods , Glioblastoma/radiotherapy , Humans , Immediate-Early Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology
3.
Oncotarget ; 6(35): 37511-25, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26462020

ABSTRACT

The SGK1 kinase is pivotal in signal transduction pathways operating in cell transformation and tumor progression. Here, we characterize in depth a novel potent and selective pyrazolo[3,4-d]pyrimidine-based SGK1 inhibitor. This compound, named SI113, active in vitro in the sub-micromolar range, inhibits SGK1-dependent signaling in cell lines in a dose- and time-dependent manner. We recently showed that SI113 slows down tumor growth and induces cell death in colon carcinoma cells, when used in monotherapy or in combination with paclitaxel. We now demonstrate for the first time that SI113 inhibits tumour growth in hepatocarcinoma models in vitro and in vivo. SI113-dependent tumor inhibition is dose- and time-dependent. In vitro and in vivo SI113-dependent SGK1 inhibition determined a dramatic increase in apoptosis/necrosis, inhibited cell proliferation and altered the cell cycle profile of treated cells. Proteome-wide biochemical studies confirmed that SI113 down-regulates the abundance of proteins downstream of SGK1 with established roles in neoplastic transformation, e.g. MDM2, NDRG1 and RAN network members. Consistent with knock-down and over-expressing cellular models for SGK1, SI113 potentiated and synergized with radiotherapy in tumor killing. No short-term toxicity was observed in treated animals during in vivo SI113 administration. These data show that direct SGK1 inhibition can be effective in hepatic cancer therapy, either alone or in combination with radiotherapy.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Gamma Rays , Gene Expression Regulation, Neoplastic/drug effects , Immediate-Early Proteins/antagonists & inhibitors , Liver Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Radiation Tolerance/drug effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Blotting, Western , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/radiotherapy , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Female , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Immediate-Early Proteins/metabolism , Immunoenzyme Techniques , In Vitro Techniques , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Liver Neoplasms/radiotherapy , Mice , Mice, Inbred NOD , Mice, SCID , Protein Serine-Threonine Kinases/metabolism , Proteome/analysis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Curr Pharm Biotechnol ; 14(13): 1099-104, 2014.
Article in English | MEDLINE | ID: mdl-24725130

ABSTRACT

Malignant glioma is a primary tumor of the central nervous system, representing a major cause of mortality in a young, productive subset of population. The management of this neoplasm requires aggressive treatments, including radiotherapy. Accurate imaging plays a central role in treatment planning process with curative intent based on radiation therapy. In order to maximize the radiation dose to the tumor and to minimize the damage to the normal surrounding tissue, a reliable identification of viable tumor margins is indeed required. The use of PET in the treatment planning process has become more promising over the years, although many important questions must be addressed. The aim of this article is to critically review the evidence supporting PET in radiotherapy planning, with special emphasis on the role of novel radiopharmaceuticals, comparing its sensitivity and specificity with respect to 18F-FDG and other anatomic imaging modalities.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain Neoplasms/radiotherapy , Glioma/diagnostic imaging , Glioma/radiotherapy , Positron-Emission Tomography/methods , Radiopharmaceuticals/therapeutic use , Humans , Patient Care Planning
5.
Cancer Biol Ther ; 15(6): 797-805, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24657936

ABSTRACT

Radiotherapy is one of the most effective therapeutic strategies for breast cancer patients, although its efficacy may be reduced by intrinsic radiation resistance of cancer cells. Recent investigations demonstrate a link between cancer cell radio-resistance and activation of sphingosine kinase (SphK1), which plays a key role in the balance of lipid signaling molecules. Sphingosine kinase (SphK1) activity can alter the sphingosine-1-phosphate (S1P)/ceramide ratio leading to an imbalance in the sphingolipid rheostat. Fingolimod (FTY720) is a novel sphingosine analog and a potent immunosuppressive drug that acts as a SphK1 antagonist, inhibits the growth, and induces apoptosis in different human cancer cell lines. We sought to investigate the in vitro radiosensitizing effects of FTY720 on the MDA-MB-361 breast cancer cell line and to assess the effects elicited by radiation and FTY720 combined treatments. We found that FTY720 significantly increased anti-proliferative and pro-apoptotic effects induced by a single dose of ionizing radiation while causing autophagosome accumulation. At the molecular level, FTY720 significantly potentiated radiation effects on perturbation of signaling pathways involved in regulation of cell cycle and apoptosis, such as PI3K/AKT and MAPK. In conclusion, our data highlight a potent radiosensitizing effect of FTY720 on breast cancer cells and provide the basis of novel therapeutic strategies for breast cancer treatment.


Subject(s)
Propylene Glycols/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Sphingosine/analogs & derivatives , Apoptosis/radiation effects , Breast Neoplasms , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/radiation effects , Drug Screening Assays, Antitumor , Female , Fingolimod Hydrochloride , Humans , Resting Phase, Cell Cycle , Sphingosine/pharmacology
6.
Oncol Lett ; 6(4): 897-900, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24137433

ABSTRACT

The aims of radiotherapeutic treatment of brain metastases include maintaining neurocognitive function and improvement of survival. Based on these premises, we present a case report in which the role of repeat stereotactic radiosurgery (SRS) was investigated in a patient with a recurrent brain metastasis from non-small cell lung cancer in the same area as previously treated with radiosurgery. A 40-year-old male caucasian patient was diagnosed with brain metastasis from non-small cell lung cancer (NSCLC) and underwent SRS. The patient developed a recurrence of the disease and a second SRS on the same area was performed. After 8 months, tumor restaging demonstrated a lesion compatible with a recurrence and the patient underwent surgery. Histological diagnosis following surgery revealed only the occurrence of radionecrosis. Radiotherapy was well-tolerated and no grade 3/4 neurological toxicity occurred. To date, no consensus exists on the efficacy of retreatment with SRS. Despite the limited number of studies in this field, in the present case report, we outline the outcomes of this unconventional approach.

SELECTION OF CITATIONS
SEARCH DETAIL
...