Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
J Contemp Brachytherapy ; 10(3): 202-210, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30038639

ABSTRACT

PURPOSE: External beam radiation therapy (EBRT) and brachytherapy (BT) with concurrent cisplatin is the standard of care for locally advanced cervical cancer. The applicability of image-guided adaptive volume-based high-dose-rate (HDR) intracavitary brachytherapy planning is an active area of investigation. In this study, we examined whether volume-based HDR-BT (HDRVOL) plans leads to more conformal plans compared to Point A (HDRPointA)-based plans. MATERIAL AND METHODS: Two hundred and forty HDRPointA plans from 48 cervical cancer patients treated with chemoradiotherapy were retrospectively collected. Point A plans were renormalized with respect to the high-risk clinical target volume (HR-CTV) for the HDRVOL plans. The doses to organs at risk (OAR; rectum, sigmoid, and bladder), and HR-CTV and the conformal index were compared between HDRPointA and HDRVOL plans. RESULTS: HDRVOL plans resulted in a 6-12% reduction in the total dose (EBRT + HDR-BT) to 0.1 cc, 1.0 cc, and 2.0 cc of the OAR as well as an 8-37% reduction in the dose to 2 cc of OAR per HDR-BT fraction compared to HDRPointA plans. Differences in the conformal indexes between the two groups of plans showed an 18-31% relative increase per HDR-BT fraction for HDRVOL plans. The D90 of the HR-CTV was reduced by 11% by HDRVOL planning and had a median dose of 86 Gy. CONCLUSIONS: Our study reports the relative improvement in OAR doses per HDR-BT fraction by HDRVOL planning compared to HDRPointA planning and demonstrates the dosimetric advantages of volume-based HDR-BT planning in creating more conformal plans.

2.
Transl Res ; 162(2): 77-92, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23567332

ABSTRACT

The role of cyclooxygenase-2 (COX-2), its lipid metabolite prostaglandin E2 (PGE2), and Eicosanoid (EP) receptors (EP; 1-4) underlying the proinflammatory mechanistic aspects of Burkitt's lymphoma, nasopharyngeal carcinoma, cervical cancer, prostate cancer, colon cancer, and Kaposi's sarcoma (KS) is an active area of investigation. The tumorigenic potential of COX-2 and PGE2 through EP receptors forms the mechanistic context underlying the chemotherapeutic potential of nonsteroidal anti-inflammatory drugs (NSAIDs). Although role of the COX-2 is described in several viral associated malignancies, the biological significance of the COX-2/PGE2/EP receptor inflammatory axis is extensively studied only in Kaposi's sarcoma-associated herpes virus (KSHV/HHV-8) associated malignancies such as KS, a multifocal endothelial cell tumor and primary effusion lymphoma (PEL), a B cell-proliferative disorder. The purpose of this review is to summarize the salient findings delineating the molecular mechanisms downstream of COX-2 involving PGE2 secretion and its autocrine and paracrine interactions with EP receptors (EP1-4), COX-2/PGE2/EP receptor signaling regulating KSHV pathogenesis and latency. KSHV infection induces COX-2, PGE2 secretion, and EP receptor activation. The resulting signal cascades modulate the expression of KSHV latency genes (latency associated nuclear antigen-1 [LANA-1] and viral-Fas (TNFRSF6)-associated via death domain like interferon converting enzyme-like- inhibitory protein [vFLIP]). vFLIP was also shown to be crucial for the maintenance of COX-2 activation. The mutually interdependent interactions between viral proteins (LANA-1/vFLIP) and COX-2/PGE2/EP receptors was shown to play key roles in the biological mechanisms involved in KS and PEL pathogenesis such as blockage of apoptosis, cell cycle regulation, transformation, proliferation, angiogenesis, adhesion, invasion, and immune-suppression. Understanding the COX-2/PGE2/EP axis is very important to develop new safer and specific therapeutic modalities for KS and PEL. In addition to COX-2 being a therapeutic target, EP receptors represent ideal targets for pharmacologic agents as PGE2 analogues and their blockers/antagonists possess antineoplastic activity, without the reported gastrointestinal and cardiovascular toxicity observed with few a NSAIDs.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Lymphoma, Primary Effusion/metabolism , Receptors, Eicosanoid/metabolism , Sarcoma, Kaposi/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Viral , Humans , Lymphoma, Primary Effusion/drug therapy , Sarcoma, Kaposi/drug therapy , Sarcoma, Kaposi/virology , Signal Transduction , Virus Latency/genetics
3.
Transl Res ; 161(6): 447-68, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23523954

ABSTRACT

The effective antitumorigenic potential of nonsteroidal anti-inflammatory drugs (NSAIDs) and eicosonoid (EP; EP1-4) receptor antagonists prompted us to test their efficacy in Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr virus (EBV) related lymphomas. Our study demonstrated that (1) EP1-4 receptor protein levels vary among the various non-Hodgkin's lymphoma (NHL) cell lines tested (BCBL-1:KSHV+/EBV-;BC-3: KSHV+/EBV-; Akata/EBV+: KSHV-/EBV+; and JSC-1 cells: KSHV+/EBV + cells); (2) 5.0 µM of EP1 antagonist (SC-51322) had a significant antiproliferative effect on BCBL-1, BC-3, Akata/EBV+, and JSC-1 cells; (3) 50.0 µM of EP2 antagonist (AH6809) was required to induce a significant antiproliferative effect on BCBL-1, Akata/EBV+, and JSC-1 cells; (4) 5.0 µM of EP4 antagonist (GW 627368X) had a significant antiproliferative effect on BC-3, Akata/EBV+, and JSC-1 cells; (5) COX-2 selective inhibitor celecoxib (5.0 µM) had significant antiproliferative effects on BCBL-1, BC-3, Akata/EBV+, and JSC-1 cells; and (6) a combination of 1.0 µM each of celecoxib, SC-51322 and GW 627368X could potentiate the proapoptotic properties of celecoxib or vice-versa. Overall, our studies identified the synergistic antiproliferative effect of NSAIDs and EP receptor blockers on KSHV and EBV related B cell malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cyclooxygenase Inhibitors/pharmacology , Lymphoma, B-Cell/pathology , Molecular Targeted Therapy/methods , Receptors, Eicosanoid/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Celecoxib , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Drug Screening Assays, Antitumor , Drug Synergism , Herpesviridae Infections/drug therapy , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Herpesvirus 4, Human/pathogenicity , Herpesvirus 8, Human/pathogenicity , Humans , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/virology , Pyrazoles/pharmacology , Receptors, Eicosanoid/metabolism , Sulfonamides/pharmacology
4.
PLoS One ; 6(9): e24379, 2011.
Article in English | MEDLINE | ID: mdl-21980345

ABSTRACT

The significance of inflammation in KSHV biology and tumorigenesis prompted us to examine the role of COX-2 in primary effusion lymphoma (PEL), an aggressive AIDS-linked KSHV-associated non-Hodgkin's lymphoma (NHL) using nimesulide, a well-known COX-2 specific NSAID. We demonstrate that (1) nimesulide is efficacious in inducing proliferation arrest in PEL (KSHV+/EBV-; BCBL-1 and BC-3, KSHV+/EBV+; JSC-1), EBV-infected (KSHV-/EBV+; Raji) and non-infected (KSHV-/EBV-; Akata, Loukes, Ramos, BJAB) high malignancy human Burkitt's lymphoma (BL) as well as KSHV-/EBV+ lymphoblastoid (LCL) cell lines; (2) nimesulide is selectively toxic to KSHV infected endothelial cells (TIVE-LTC) compared to TIVE and primary endothelial cells (HMVEC-d); (3) nimesulide reduced KSHV latent gene expression, disrupted p53-LANA-1 protein complexes, and activated the p53/p21 tumor-suppressor pathway; (4) COX-2 inhibition down-regulated cell survival kinases (p-Akt and p-GSK-3ß), an angiogenic factor (VEGF-C), PEL defining genes (syndecan-1, aquaporin-3, and vitamin-D3 receptor) and cell cycle proteins such as cyclins E/A and cdc25C; (5) nimesulide induced sustained cell death and G1 arrest in BCBL-1 cells; (6) nimesulide substantially reduced the colony forming capacity of BCBL-1 cells. Overall, our studies provide a comprehensive molecular framework linking COX-2 with PEL pathogenesis and identify the chemotherapeutic potential of nimesulide in treating PEL.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Herpesvirus 8, Human/metabolism , Lymphoma, Primary Effusion/drug therapy , Sulfonamides/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Separation , Cell Survival , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Time Factors , Tumor Suppressor Protein p53/metabolism
5.
Virology ; 406(1): 103-16, 2010 Oct 10.
Article in English | MEDLINE | ID: mdl-20674951

ABSTRACT

KSHV effectively binds, enters and establishes infection in THP-1 cells with initial concurrent expression of latent ORF73 and lytic ORF50 genes and subsequent persistence of ORF73. KSHV genome persisted for 30 days and lytic cycle could be activated. KSHV utilized heparan sulfate for binding to THP-1 cells and primary monocytes. Blocking DC-SIGN did not inhibit KSHV binding; however, virus entry in THP-1 cells and in primary monocytes was reduced. In addition to the previously identified integrins alpha3beta1, alphavbeta3 and alphavbeta5, integrin alpha5beta1 was also utilized for infection. KSHV entered THP-1 cells via clathrin and caveolin mediated endocytosis and did not utilize macropinocytosis as in human dermal endothelial cells, and required an endosomal acidification. Infection also induced phosphorylation of FAK, Src, PI3K, NF-kappaB and ERK1/2 signaling molecules, and entry was blocked by tyrosine kinase inhibitors. These findings suggest that THP-1 cells are highly useful model for studying KSHV infection of monocytes.


Subject(s)
Cell Adhesion Molecules/physiology , Heparitin Sulfate/physiology , Herpesvirus 8, Human/physiology , Herpesvirus 8, Human/pathogenicity , Integrins/physiology , Lectins, C-Type/physiology , Monocytes/virology , Receptors, Cell Surface/physiology , Antigens, Viral/genetics , Cell Line , Cell Nucleus/virology , DNA, Viral/genetics , DNA, Viral/metabolism , Endocytosis/physiology , Gene Expression , Herpesvirus 8, Human/genetics , Host-Pathogen Interactions/physiology , Humans , Immediate-Early Proteins/genetics , Kinetics , Monocytes/physiology , Nuclear Proteins/genetics , Signal Transduction/physiology , Trans-Activators/genetics , Virus Internalization
6.
PLoS Pathog ; 6(2): e1000777, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20169190

ABSTRACT

Kaposi's sarcoma (KS), an enigmatic endothelial cell vascular neoplasm, is characterized by the proliferation of spindle shaped endothelial cells, inflammatory cytokines (ICs), growth factors (GFs) and angiogenic factors. KSHV is etiologically linked to KS and expresses its latent genes in KS lesion endothelial cells. Primary infection of human micro vascular endothelial cells (HMVEC-d) results in the establishment of latent infection and reprogramming of host genes, and cyclooxygenase-2 (COX-2) is one of the highly up-regulated genes. Our previous study suggested a role for COX-2 in the establishment and maintenance of KSHV latency. Here, we examined the role of COX-2 in the induction of ICs, GFs, angiogenesis and invasive events occurring during KSHV de novo infection of endothelial cells. A significant amount of COX-2 was detected in KS tissue sections. Telomerase-immortalized human umbilical vein endothelial cells supporting KSHV stable latency (TIVE-LTC) expressed elevated levels of functional COX-2 and microsomal PGE2 synthase (m-PGES), and secreted the predominant eicosanoid inflammatory metabolite PGE2. Infected HMVEC-d and TIVE-LTC cells secreted a variety of ICs, GFs, angiogenic factors and matrix metalloproteinases (MMPs), which were significantly abrogated by COX-2 inhibition either by chemical inhibitors or by siRNA. The ability of these factors to induce tube formation of uninfected endothelial cells was also inhibited. PGE2, secreted early during KSHV infection, profoundly increased the adhesion of uninfected endothelial cells to fibronectin by activating the small G protein Rac1. COX-2 inhibition considerably reduced KSHV latent ORF73 gene expression and survival of TIVE-LTC cells. Collectively, these studies underscore the pivotal role of KSHV induced COX-2/PGE2 in creating KS lesion like microenvironment during de novo infection. Since COX-2 plays multiple roles in KSHV latent gene expression, which themselves are powerful mediators of cytokine induction, anti-apoptosis, cell survival and viral genome maintainence, effective inhibition of COX-2 via well-characterized clinically approved COX-2 inhibitors could potentially be used in treatment to control latent KSHV infection and ameliorate KS.


Subject(s)
Cyclooxygenase 2/metabolism , Herpesvirus 8, Human/physiology , Inflammation/virology , Neovascularization, Pathologic/virology , Sarcoma, Kaposi/enzymology , Virus Latency/physiology , Blotting, Western , Cell Adhesion/physiology , Cell Separation , Endothelial Cells/metabolism , Endothelial Cells/virology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunohistochemistry , Inflammation/enzymology , Neovascularization, Pathologic/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Kaposi/virology
7.
Virology ; 392(1): 34-51, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19640558

ABSTRACT

KSHV vGPCR, a lytic cycle associated protein, induces several signaling pathways leading to the activation of various transcription factors and consequently the expression of cellular and viral genes. Though the role of vGPCR in KSHV tumorigenicity has been well studied, its function related to the viral life cycle is poorly understood. Reduction in vGPCR by RNA interference also resulted in the reduction in KSHV lytic switch ORF50 gene and protein expression. Induction of vGPCR by doxycycline in BC3.14 cells also resulted in more KSHV production. When this was explored, induction of the ORF50 promoter by vGPCR expression was observed. Further examination of the molecular mechanisms by which vGPCR regulates the ORF50 promoter, using various ORF50 promoter constructs, revealed that induction of ORF50 promoter by vGPCR did not involve AP1 but was dependent on Sp1 and Sp3 transcription factors. vGPCR signaling led to an increase in Sp1 and Sp3 DNA binding activity and a decrease in histone deacetylase (HDAC) activity. These activities were pertussis toxin independent, did not involve Rho and Rac-GTPases and involved the heterotrimeric G protein subunits Galpha12 and Galphaq. Studies using pharmacologic inhibitors and dominant-negative proteins identified phospholipase C, the novel protein kinase C (novel PKC) family and protein kinase D (PKD) as part of the signaling initiated by vGPCR leading to ORF50 promoter activation. Taken together, this study suggests a role for vGPCR in the sustained expression of ORF50 which could lead to a continued activation of lytic cycle genes and ultimately to successful viral progeny formation.


Subject(s)
Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/physiology , Immediate-Early Proteins/genetics , Immediate-Early Proteins/physiology , Receptors, Chemokine/genetics , Receptors, Chemokine/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Base Sequence , Binding Sites , Cell Line , Doxycycline/pharmacology , Feedback, Physiological , Gene Expression/drug effects , Genes, Viral , Herpesvirus 8, Human/pathogenicity , Humans , Models, Biological , Mutation , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Signal Transduction , Sp1 Transcription Factor/metabolism , rho GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL