Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Cancer Rep (Hoboken) ; 6(2): e1733, 2023 02.
Article in English | MEDLINE | ID: mdl-36241419

ABSTRACT

BACKGROUND: Melanoma is one of the most aggressive cancers, with 1.6% of total cancer deaths in the United States. In recent years treatment options for metastatic melanoma have been improved by the FDA approval of new therapeutic agents. However, these inhibitors-based therapies are non-specific and have severe toxicities, including hyperkeratosis, photosensitivity, hepatitis, arthralgia, and fatigue. AIMS: The aim of this study is to determine the synthetic lethal effect (paclitaxel and radiations) on melanoma cells and reduce the total radiation doses by increasing the dose rates up to 2400 MU/min. METHODS AND RESULTS: We previously reported a radiation treatment (10 MV x-rays, 10X-FFF, dose rate 2400MU/min, low total dose 0.5 Gy) that kills melanoma cells with 80% survival of normal HEM in vitro. In this study, we extended the radiation cycle up to four and included paclitaxel treatment to study the synthetic lethal effect on melanoma and two other normal primary cells, HDF and HEK. Cells were treated with paclitaxel prior to the radiation at a dose rate of 400 and 2400 MU/min with a total radiation dose of only 0.5 Gy. Mitochondrial respiration assay, DNA damage assay, and colony formation assays were performed to study apoptosis and cell death induction. Four days of consequent radiation treatment with paclitaxel significantly reduces the survival of melanoma cells by inducing apoptosis and mitochondrial damage. After treatment, excessive DNA damage in melanoma cells leads to an increase in the expression of pro-apoptotic genes (Caspase-3) and a decrease in the expression of DNA repair gene (PARP1) and anti-apoptotic gene (Bcl-2) to activate the apoptosis pathway. The combination of paclitaxel and radiation reduces the survival of melanoma cells colonies compared to radiation alone. CONCLUSION: Our study indicates that radiations with paclitaxel have a potential synthetic lethal effect on melanoma cells and can be developed as a melanoma therapy without toxicities or harmful effects on normal primary skin cells.


Subject(s)
Melanoma , Paclitaxel , Humans , X-Rays , Melanoma/drug therapy , Apoptosis
2.
Cancer Rep (Hoboken) ; 4(2): e1311, 2021 04.
Article in English | MEDLINE | ID: mdl-33103852

ABSTRACT

BACKGROUND: Classical Hodgkin lymphoma (cHL) is a unique lymphoid malignancy with a tumor microenvironment (TME) consisting of a small number of neoplastic-Hodgkin and Reed-Sternberg (H-RS) cells (<1%), surrounded by a large number of nonneoplastic infiltrating immune cells (>90%). The TME of cHL critically depends on immune cells to support tumor growth as H-RS cells cannot survive and proliferate in isolation. RECENT FINDINGS: Programmed cell death protein 1 (PD-1) ligand expressed on H-RS cells inhibits the clearance of tumor by causing T-cell exhaustion. Nivolumab and pembrolizumab, PD-1 inhibitors, have been proven to be effective in treating adult and pediatric patients with R/R cHL. Tumor-associated macrophages (TAMs) are a central component of TME and are known to cause poor prognosis in adult HL. However, the prognostic impact of CD68+ TAMs in pediatric HL remains ambiguous. EBV modulates the tumor milieu of HL and plays a strategic role in immune escape by enrichment of the TME with Treg cells and associated immunosuppressive cytokines in adult HL. In contrast, EBV+ pediatric patients have increased infiltration of CD8+ T-cells and show a better therapeutic response suggesting viral-related TME is distinct in childhood HL. The role of CASP3 in apoptosis of H-RS cells and its correlation with response prediction in adult and pediatric HL suggest it may serve as a potential biomarker. In cHL, CD30, EBV, and NF-κB signaling employ exosomes for cell-cell communication that triggers the migration capacity of fibroblasts, stimulate to produce proinflammatory cytokines, and help to create a tumor-supportive microenvironment. CONCLUSION: The cHL microenvironment is distinct in adult and pediatric HL. Future studies are required to understand the role of interplay between H-RS cells and EBV-associated microenvironment and their clinical outcome. They may present novel therapeutic targets for the development of antilymphoma therapy.


Subject(s)
Cytokines/metabolism , Epstein-Barr Virus Infections/immunology , Hodgkin Disease/immunology , Reed-Sternberg Cells/pathology , Tumor Microenvironment/immunology , Adult , Age Factors , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Caspase 3/metabolism , Cell Communication/immunology , Child , Epstein-Barr Virus Infections/microbiology , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Exosomes/metabolism , Herpesvirus 4, Human/immunology , Hodgkin Disease/mortality , Hodgkin Disease/pathology , Hodgkin Disease/virology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Prognosis , Programmed Cell Death 1 Receptor/metabolism , Reed-Sternberg Cells/immunology , Tumor Escape , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
3.
Oncotarget ; 11(44): 4045-4073, 2020 Nov 03.
Article in English | MEDLINE | ID: mdl-33216822

ABSTRACT

Despite the great efforts for better treatment options for diffuse large B-cell lymphoma (DLBCL) (most common form of non-Hodgkin lymphoma, NHL) to treat and prevent relapse, it continues to be a challenge. Here, we present an overview of DLBCL and address the diagnostic assays and molecular techniques used in its diagnosis, role of biomarkers in detection, treatment of early and advanced stage DLBCL, and novel drug regimens. We discuss the significant biomarkers that have emerged as essential tools for stratifying patients according to risk factors and for providing insights into the use of more targeted and individualized therapeutics. We discuss techniques such as gene expression studies, including next-generation sequencing, which have enabled a more understanding of the complex pathogenesis of DLBCL and have helped determine molecular targets for novel therapeutic agents. We examine current treatment approaches, outline the findings of completed clinical trials, and provide updates for ongoing clinical trials. We highlight clinical trials relevant to the significant fraction of DLBCL patients who present with complex cases marked by high relapse rates. Supported by an increased understanding of targetable pathways in DLBCL, clinical trials involving specialized combination therapies are bringing us within reach the promise of an effective cure to DLBCL using precision medicine. Optimization of therapy remains a crucial objective, with the end goal being a balance between high survival rates through targeted and personalized treatment while reducing adverse effects in DLBCL patients of all subsets.

4.
Oncotarget ; 7(41): 67551-67573, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27563824

ABSTRACT

Hodgkin lymphoma (HL) is a lymphoid malignancy that is typically derived from germinal-center B cells. EBV infection, mutations in NF-κB pathway genes, and genetic susceptibility are known risk factors for developing HL. CD30 and NF-κB have been identified as potential biomarkers in pediatric HL patients, and these molecules may represent therapeutic targets. Although current risk adapted and response based treatment approaches yield overall survival rates of >95%, treatment of relapse or refractory patients remains challenging. Targeted HL therapy with the antibody-drug conjugate Brentuximab vedotin (Bv) has proven to be superior to conventional salvage chemotherapy and clinical trials are being conducted to incorporate Bv into frontline therapy that substitutes Bv for alkylating agents to minimize secondary malignancies. The appearance of secondary malignancies has been a concern in pediatric HL, as these patients are at highest risk among all childhood cancer survivors. The risk of developing secondary leukemia following childhood HL treatment is 10.4 to 174.8 times greater than the risk in the general pediatric population and the prognosis is significantly poorer than the other hematological malignancies with a mortality rate of nearly 100%. Therefore, identifying clinically valuable biomarkers is of utmost importance to stratify and select patients who may or may not need intensive regimens to maintain optimal balance between maximal survival rates and averting late effects. Here we discuss epidemiology, risk factors, staging, molecular and genetic prognostic biomarkers, treatment for low and high-risk patients, and the late occurrence of secondary malignancies in pediatric HL.


Subject(s)
Hodgkin Disease , Adolescent , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Child , Child, Preschool , Clinical Trials as Topic , Female , Humans , Male , Translational Research, Biomedical
5.
Oncotarget ; 7(28): 44735-44762, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27007053

ABSTRACT

Fibroblast growth factor (FGF) signaling is essential for normal and cancer biology. Mammalian FGF family members participate in multiple signaling pathways by binding to heparan sulfate and FGF receptors (FGFR) with varying affinities. FGF2 is the prototype member of the FGF family and interacts with its receptor to mediate receptor dimerization, phosphorylation, and activation of signaling pathways, such as Ras-MAPK and PI3K pathways. Excessive mitogenic signaling through the FGF/FGFR axis may induce carcinogenic effects by promoting cancer progression and increasing the angiogenic potential, which can lead to metastatic tumor phenotypes. Dysregulated FGF/FGFR signaling is associated with aggressive cancer phenotypes, enhanced chemotherapy resistance and poor clinical outcomes. In vitro experimental settings have indicated that extracellular FGF2 affects proliferation, drug sensitivity, and apoptosis of cancer cells. Therapeutically targeting FGF2 and FGFR has been extensively assessed in multiple preclinical studies and numerous drugs and treatment options have been tested in clinical trials. Diagnostic assays are used to quantify FGF2, FGFRs, and downstream signaling molecules to better select a target patient population for higher efficacy of cancer therapies. This review focuses on the prognostic significance of FGF2 in cancer with emphasis on therapeutic intervention strategies for solid and hematological malignancies.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Hematologic Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Precision Medicine/methods , Animals , Antineoplastic Agents/therapeutic use , Fibroblast Growth Factor 2/antagonists & inhibitors , Hematologic Neoplasms/drug therapy , Humans , Molecular Targeted Therapy/methods , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction/drug effects
6.
Oncotarget ; 6(30): 28693-715, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26293675

ABSTRACT

Syndecan-1 (SDC1, CD138) is a key cell surface adhesion molecule essential for maintaining cell morphology and interaction with the surrounding microenvironment. Deregulation of SDC1 contributes to cancer progression by promoting cell proliferation, metastasis, invasion and angiogenesis, and is associated with relapse through chemoresistance. SDC1 expression level is also associated with responses to chemotherapy and with prognosis in multiple solid and hematological cancers, including multiple myeloma and Hodgkin lymphoma. At the tissue level, the expression levels of SDC1 and the released extracellular domain of SDC1 correlate with tumor malignancy, phenotype, and metastatic potential for both solid and hematological tumors in a tissue-specific manner. The SDC1 expression profile varies among cancer types, but the differential expression signatures between normal and cancer cells in epithelial and stromal compartments are directly associated with aggressiveness of tumors and patient's clinical outcome and survival. Therefore, relevant biomarkers of SDC signaling may be useful for selecting patients that would most likely respond to a particular therapy at the time of diagnosis or perhaps for predicting relapse. In addition, the reciprocal expression signature of SDC between tumor epithelial and stromal compartments may have synergistic value for patient selection and the prediction of clinical outcome.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Syndecan-1/metabolism , Animals , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Prognosis , Signal Transduction , Syndecan-1/genetics , Tumor Microenvironment
7.
J Control Release ; 188: 44-52, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-24928321

ABSTRACT

Cell-based delivery of cell penetrating peptides (CPPs) could represent a new platform for intracellular peptide delivery to local tissues. Expressed CPPs, coupled to a secretory signal peptide (SP), can support intercellular transport. However, low secretion efficiency, which may correlate with the positive charge of most CPPs, has emerged as one of the main impediments for efficient intercellular transport. We have reported that a modified Tat-based CPP (Tatm) with reduced positive charge is secreted efficiently, but its transduction activity was greatly reduced. We now show that a triple repeat of Tatm (Tatm3x) with an elongated α-helical amphipathic structure enhances transduction activity and simultaneously retains its secretion efficacy, although passage through the secretory pathway reduces its cell-penetrating activity. SP-Tatm3x supports intercellular transport of fused fluorescent proteins, as well as cell entry and function of a pro-apoptotic peptide. In addition, SP-Tatm3x largely escapes RNA inhibition, which is identified as another potential impediment to CPP-mediated intercellular transport. Expression of SP-Tatm3x in heparan sulfate proteoglycan-negative cells further improves its transduction activity. These results demonstrate the feasibility of intercellular transport of proteins, but further work is needed to better understand the reduction of cell-penetrating activity associated with secretion of CPP-fusion proteins.


Subject(s)
Cell-Penetrating Peptides/metabolism , Green Fluorescent Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Adenoviridae/genetics , Amino Acid Sequence , Cell Line , Cell-Penetrating Peptides/analysis , Cell-Penetrating Peptides/genetics , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Molecular Sequence Data , Plasmids/chemistry , Plasmids/genetics , Protein Sorting Signals , Protein Transport , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Transfection
8.
Ann Clin Microbiol Antimicrob ; 9: 24, 2010 Sep 08.
Article in English | MEDLINE | ID: mdl-20822551

ABSTRACT

BACKGROUND: Screening women for Chlamydia trachomatis infection in developing countries is highly desirable because of asymptomatic infection. The existing diagnostic methods in developing countries are not effective and their sensitivity fall below 45.0% which leads to further spread of infection. There is an urgent need for improved and cost effective diagnostic tests that will reduce the burden of sexually transmitted infections in the developing world. METHODS: Prevalence of C. trachomatis infection among women visiting gynaecology department of Hindu Rao hospital in Delhi, India was determined using Roche Amplicor Multi Well Plate kit (MWP) as well as using in-house PCR assay. We used 593 endocervical swabs for clinical evaluation of the in-house developed assay against Direct Fluorescence Assay (DFA; Group I n = 274) and Roche Amplicor MWP kit (Group II, n = 319 samples) and determined the sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV) of the in-house developed assay. RESULTS: We detected 23.0% positive cases and there was a higher representation of women aged 18-33 in this group. An in-house PCR assay was developed and evaluated by targeting unique sequence within the gyrA gene of C. trachomatis. Specificity of the reaction was confirmed by using genomic DNA of human and other STI related microorganisms as template. Assay is highly sensitive and can detect as low as 10 fg of C. trachomatis DNA. The resolved sensitivity of in-house PCR was 94.5% compared with 88.0% of DFA assay. The high specificity (98.4%) and sensitivity (97.1%) of the in-house assay against Roche kit and availability of test results within 3 hours allowed for immediate treatment and reduced the risk of potential onward transmission. CONCLUSIONS: The in-house PCR method is cost effective (~ 20.0% of Roche assay) and hence could be a better alternative for routine diagnosis of genital infection by C. trachomatis to facilitate improved screening and treatment management.


Subject(s)
Chlamydia Infections/diagnosis , Chlamydia trachomatis/isolation & purification , Genital Diseases, Female/diagnosis , Outpatient Clinics, Hospital , Polymerase Chain Reaction/methods , Pregnancy Complications, Infectious/diagnosis , Adult , Chlamydia Infections/epidemiology , DNA, Bacterial/analysis , Developing Countries , Female , Genital Diseases, Female/epidemiology , Humans , In Situ Hybridization, Fluorescence , India/epidemiology , Male , Middle Aged , Predictive Value of Tests , Pregnancy , Pregnancy Complications, Infectious/epidemiology , Reagent Kits, Diagnostic , Sensitivity and Specificity , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...