Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Vaccines (Basel) ; 11(7)2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37515009

ABSTRACT

Cancer patients on chemotherapy have a lower immune response to SARS-CoV-2 vaccines. Therefore, through a prospective cohort study of patients with solid tumors receiving chemotherapy, we aimed to determine the immunogenicity of an mRNA vaccine booster (BNT162b2) among patients previously immunized with an inactivated (CoronaVac) or homologous (BNT162b2) SARS-CoV-2 vaccine. The primary outcome was the proportion of patients with anti-SARS-CoV-2 neutralizing antibody (NAb) seropositivity at 8-12 weeks post-booster. The secondary end points included IgG antibody (TAb) seropositivity and specific T-cell responses. A total of 109 patients were included. Eighty-four (77%) had heterologous vaccine schedules (two doses of CoronaVac followed by the BNT162b2 booster) and twenty-five had (23%) homologous vaccine schedules (three doses of BNT162b2). IgG antibody positivity for the homologous and heterologous regimen were 100% and 96% (p = 0.338), whereas NAb positivity reached 100% and 92% (p = 0.13), respectively. Absolute NAb positivity and Tab levels were associated with the homologous schedule (with a beta coefficient of 0.26 with p = 0.027 and a geometric mean ratio 1.41 with p = 0.044, respectively). Both the homologous and heterologous vaccine regimens elicited a strong humoral and cellular response after the BNT162b2 booster. The homologous regimen was associated with higher NAb positivity and Tab levels after adjusting for relevant covariates.

2.
Oncotarget ; 8(14): 23073-23086, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28160570

ABSTRACT

The interaction between acute myeloid leukemia cells (AML) with the bone marrow stroma cells (BMSCs) determines a protective environment that favors tumor development and resistance to conventional chemotherapy. We showed that BMSCs secrete soluble factors that protect AML cells from Ara-C induced cytotoxicity. This leukemia chemoresistance is associated with a decrease in the equilibrative nucleoside transporter (ENT1) activity by inducing removal of ENT1 from the cell surface. Reduction of cell proliferation was also observed with activation of AKT and mTOR-dependent cell survival pathways, which may also contribute to the tumor chemoprotection. Analysis of primary BMSC cultures has demonstrated that AML patients with stroma capable to confer Ara-C resistance in vitro compared to AML patients without this stroma capacity were associated with a worse prognosis. The two year overall survival rate was 0% versus 80% respectively (p=0.0001). This is the first report of a chemoprotection mechanism based on the removal of a drug transporter from the cell surface and most importantly the first time that a stroma phenotype has correlated with prognostic outcome in cancer.


Subject(s)
Bone Marrow/metabolism , Cytarabine/pharmacology , Equilibrative Nucleoside Transporter 1/metabolism , Leukemia, Myeloid, Acute/drug therapy , Bone Marrow Cells/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Patient Outcome Assessment , Stromal Cells/pathology
3.
PLoS One ; 12(1): e0168888, 2017.
Article in English | MEDLINE | ID: mdl-28045930

ABSTRACT

Acute myeloid leukemia (AML) has a high mortality rate despite chemotherapy and transplantation. Both CXCR4/SDF-1 and VLA-4/VCAM1 axes are involved in leukemia protection but little is known about the role of CCL2/CCR2 in AML biology and protection against chemotherapy. We measured CCR2 expression in AML cell lines and primary AML cells by flow cytometry (FCM), real time PCR (RT-PCR) and western blot (WB). CCL2 production was quantified by solid phase ELISA in peripheral blood (PB) and bone marrow (BM) serum. We measured chemotaxis in a transwell system with different concentrations of CCL2/CCR2 blockers; cell cycle with BrDU and propidium iodide and proliferation with yellow tetrazolium MTT. We determined synergy in in vitro cell apoptosis combining chemotherapy and CCL2/CCR2 blockade. Finally, we performed chemoprotection studies in an in vivo mouse model. Of 35 patients, 23 (65%) expressed CCR2 by FCM in PB. Two cell lines expressed high levels of CCR2 (THP-1 and murine AML). RT-PCR and WB confirmed CCR2 production. CCL2 solid phase ELISA showed significantly lower levels of CCL2 in PB and BM compared to normal controls. Chemotaxis experiments confirmed a dose-dependent migration in AML primary cells expressing CCR2 and THP-1 cells. A significant inhibition of transmigration was seen after CCL2/CCR2 blockade. Proliferation of CCR2+ AML cell lines was slightly increased (1.4-fold) after axis stimulation. We observed a non-significant increase in phase S THP-1 cells exposed to CCL2 and a concomitant decrease of cells in G1. The chemotherapy studies did not show a protective effect of CCL2 on cytarabine-induced apoptosis or synergy with chemotherapy after CCL2/CCR2 blockade both in vitro and in vivo. In conclusion, CCL2/CCR2 axis is expressed in the majority of monocytoid AML blasts. The axis is involved in cell trafficking and proliferation but no in vitro and in vivo chemotherapy protective effect was seen.


Subject(s)
Chemokine CCL2/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Promyelocytic, Acute/drug therapy , Receptors, CCR2/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Bone Marrow/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colony-Forming Units Assay , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Monocytes/cytology , U937 Cells , Young Adult
4.
PLoS One ; 7(5): e37203, 2012.
Article in English | MEDLINE | ID: mdl-22629369

ABSTRACT

BACKGROUND: Despite a high response rate to chemotherapy, the majority of patients with acute myeloid leukemia (AML) are destined to relapse due to residual disease in the bone marrow (BM). The tumor microenvironment is increasingly being recognized as a critical factor in mediating cancer cell survival and drug resistance. In this study, we propose to identify mechanisms involved in the chemoprotection conferred by the BM stroma to leukemia cells. METHODS: Using a leukemia mouse model and a human leukemia cell line, we studied the interaction of leukemia cells with the BM microenvironment. We evaluated in vivo and in vitro leukemia cell chemoprotection to different cytotoxic agents mediated by the BM stroma. Leukemia cell apoptosis was assessed by flow cytometry and western blotting. The activity of the equilibrative nucleoside transporter 1 (ENT1), responsible for cytarabine cell incorporation, was investigated by measuring transport and intracellular accumulation of (3)H-adenosine. RESULTS: Leukemia cell mobilization from the bone marrow into peripheral blood in vivo using a CXCR4 inhibitor induced chemo-sensitization of leukemia cells to cytarabine, which translated into a prolonged survival advantage in our mouse leukemia model. In vitro, the BM stromal cells secreted a soluble factor that mediated significant chemoprotection to leukemia cells from cytarabine induced apoptosis. Furthermore, the BM stromal cell supernatant induced a 50% reduction of the ENT1 activity in leukemia cells, reducing the incorporation of cytarabine. No protection was observed when radiation or other cytotoxic agents such as etoposide, cisplatin and 5-fluorouracil were used. CONCLUSION: The BM stroma secretes a soluble factor that significantly protects leukemia cells from cytarabine-induced apoptosis and blocks ENT1 activity. Strategies that modify the chemo-protective effects mediated by the BM microenvironment may enhance the benefit of conventional chemotherapy for patients with AML.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Bone Marrow Cells/metabolism , Cytarabine/pharmacology , Equilibrative Nucleoside Transporter 1/metabolism , Leukemia, Myeloid, Acute/drug therapy , Animals , Antimetabolites, Antineoplastic/therapeutic use , Apoptosis/physiology , Bone Marrow Cells/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytarabine/therapeutic use , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Cells, Cultured
5.
Anticancer Res ; 29(5): 1483-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19443354

ABSTRACT

UNLABELLED: Cyclooxygenase-2 (COX-2) is associated with tumour promotion, inhibition of apoptosis, angiogenesis and metastasis. Celecoxib, a selective COX-2 inhibitor was investigated, in patients with clinically localized prostate cancer using immunohistochemistry. PATIENTS AND METHODS: Patients with cT1-2 prostate cancer (n=45) were randomized to celecoxib 400mg b.d. or no treatment for four weeks prior to radical prostatectomy. Histological sections of preoperative biopsy and matched radical prostatectomy specimens were stained for markers of cell proliferation (MIB-1/Ki-67), microvessel density (CD-31 with Weidner scoring), COX-2, apoptosis (TUNEL analysis), angiogenic factors (VEGF and KDR) and HIF-1. RESULTS: Celecoxib decreased tumour cell proliferation, microvessel density, angiogenesis and HIF-1 whilst enhancing apoptosis. These effects approached statistical significance in a multivariate model and the cell proliferation index approached statistical significance on univariate analysis. CONCLUSION: In this pilot study a 4 week regimen of celecoxib resulted in measurable biological effects in prostate cancer tissue. These findings warrant further investigation.


Subject(s)
Cyclooxygenase Inhibitors/therapeutic use , Prostatic Neoplasms/drug therapy , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Celecoxib , Cyclooxygenase Inhibitors/adverse effects , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Pyrazoles/adverse effects , Single-Blind Method , Sulfonamides/adverse effects
6.
J Pharmacol Exp Ther ; 324(3): 1181-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18089846

ABSTRACT

Cyclooxygenase (COX)-2 has emerged as an exciting target for therapeutic intervention in the management of cancer. Immunohistochemistry studies have indicated higher expression of COX-2 in cancerous versus benign prostatic tissue. We have explored the role of COX-2 in prostate cancer in terms of attenuation of apoptosis and sensitivity to pharmacological agents, including COX-2 inhibitors. The human prostate cancer cell line LNCaP was stably transfected with COX-2 (LNCaPCOX-2) and compared with the empty vector control line (LNCaPneo). Chemosensitivity testing indicated no change in sensitivity to the cytotoxic effects of COX-2 inhibitors celecoxib or sulindac or VP16. However, LNCaPCOX-2 cells showed 3-fold resistance to carboplatin, which was partially reversed by coincubation with the phosphatidylinositol 3-kinase inhibitor wortmannin. Concomitant with reduced apoptotic response to cytotoxic agents, LNCaPCOX-2 cells expressed increased levels of survivin and Bcl-2 with enhanced activation of AKT. We also investigated the effects of celecoxib on expression levels of genes relevant to prostate cancer and drug resistance in our model system using quantitative polymerase chain reaction analysis. Celecoxib treatment resulted in highly significant increases in the mRNA expression of the smooth muscle component desmin, the detoxification enzyme glutathione S-transferase pi (GSTpi), and nonsteroidal anti-inflammatory response gene (NAG-1) in the LNCaPCOX-2 cell line compared with LNCaPneo cells. Significant decreases in survivin levels and increases in GSTpi and NAG-1 appeared to be COX-2-dependent effects because they were more pronounced in LNCaPCOX-2 cells. Our findings indicate both COX-2-dependent and -independent mechanisms attributable to celecoxib and support its utility in the management of prostate cancer.


Subject(s)
Cyclooxygenase 2/biosynthesis , Cytotoxins/therapeutic use , Gene Expression Regulation, Neoplastic/physiology , Prostatic Neoplasms/enzymology , Celecoxib , Cell Line, Tumor , Cyclooxygenase 2/genetics , Cytotoxins/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Prostatic Neoplasms/drug therapy , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
7.
J Pharmacol Exp Ther ; 313(2): 780-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15665138

ABSTRACT

The mild analgesic drug acetaminophen (AAP) induces severe hepatic injury when taken at excessive doses. Recent evidence shows that the initial form of damage is through apoptosis, but this fails to go to completion and degenerates into necrosis. The aim of this study was to elucidate the mechanism through which AAP induces apoptosis using human HuH7 hepatoma cells as an in vitro model system to investigate the initial phase of AAP-induced hepatic injury. AAP-induced apoptosis in HuH7 cells as evidenced by chromatin condensation was preceded by the translocation of Bax to mitochondria and the cytoplasmic release of the proapoptotic factors cytochrome c and Smac/DIABLO. A concomitant loss of mitochondrial membrane potential occurred. Activation of the mitochondrial pathway of apoptosis led to the activation of execution caspases-3 and -7. AAP-induced apoptosis and cell death was blocked by inhibitors of caspases but not by inhibitors of calpains, cathepsins, and serine proteases. Apoptosis was unaffected by inhibitors of the mitochondrial permeability transition pore and by inhibitors of Jun NH(2)-terminal kinases, p38 mitogen-activated protein kinase, or mitogen-activated protein kinase kinase 1/2. However, pharmacological inhibition of glycogen synthase kinase-3 (GSK-3) delayed and decreased the extent of AAP-induced apoptosis. In comparison, endoplasmic reticulum stress-induced but not prooxidant-induced apoptosis of HuH7 cells was sensitive to GSK-3 inhibition. It is concluded that AAP-induced apoptosis involves the mitochondrial pathway of apoptosis that is mediated by GSK-3 and most likely initiated through an endoplasmic reticulum stress response.


Subject(s)
Acetaminophen/toxicity , Apoptosis/physiology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Glycogen Synthase Kinase 3/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Protein Kinase Inhibitors/pharmacology
8.
Toxicol Appl Pharmacol ; 191(2): 118-29, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12946648

ABSTRACT

The role of apoptosis in acetaminophen (AAP)-induced hepatic injury was investigated. Six hours after AAP administration to BALB/c mice, a significant loss of hepatic mitochondrial cytochrome c was observed that was similar in extent to the loss observed after in vivo activation of CD95 by antibody treatment. AAP-induced loss of mitochondrial cytochrome c coincided with the appearance in the cytosol of a fragment corresponding to truncated Bid (tBid). At the same time, tBid became detectable in the mitochondrial fraction, and concomitantly, Bax was found translocated to mitochondria. However, AAP failed to activate the execution caspases 3 and 7 as evidenced by a lack of procaspase processing and the absence of an increase in caspase-3-like activity. In contrast, the administration of the pan-inhibitor of caspases, benzyloxycarbonyl-Val-Ala-DL-Asp-fluoromethylketone (but not its analogue benzyloxycarbonyl-Phe-Ala-fluoromethylketone) prevented the development of liver injury by AAP and the appearance of apoptotic parenchymal cells. This correlated with the inhibition of the processing of Bid to tBid. The caspase inhibitor failed to prevent both the redistribution of Bax to the mitochondria and the loss of cytochrome c. In conclusion, apoptosis is an important causal event in the initiation of the hepatic injury inflicted by AAP. However, as suggested by the lack of activation of the main execution caspases, apoptosis is not properly executed and degenerates into necrosis.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Apoptosis/drug effects , Mitochondria, Liver/drug effects , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Amino Acid Chloromethyl Ketones/pharmacology , Animals , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/metabolism , Caspase Inhibitors , Caspases/metabolism , Cytochrome c Group/metabolism , Enzyme Inhibitors/pharmacology , Injections, Intraperitoneal , Male , Mice , Mice, Inbred BALB C , Mitochondria, Liver/enzymology , Mitochondria, Liver/metabolism , Proto-Oncogene Proteins/metabolism , bcl-2-Associated X Protein
9.
Ann N Y Acad Sci ; 1010: 557-9, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15033790

ABSTRACT

Apoptosis plays a critical role in acetaminophen (AAP)-induced hepatic injury, since inhibiting apoptosis also prevents the development of acute liver failure. In this study, the mechanism of apoptosis induction by AAP was investigated in the human hepatoblastoma cell line HuH7. AAP caused marked cytotoxicity in HuH7 cells as a result of apoptosis. Processing of execution caspases to their corresponding active fragments and cleavage of cytokeratin-18 were observed, supporting a role of caspases in AAP-induced apoptosis. The manifestation of apoptosis was preceded by a translocation of cytochrome c from mitochondria to the cytosol. In conclusion, AAP induces apoptosis in human hepatoblastoma HuH7 cells through mitochondrial cytochrome c release and caspase activation.


Subject(s)
Acetaminophen/toxicity , Apoptosis/drug effects , Liver/pathology , Carcinoma, Hepatocellular , Cell Line, Tumor , Cell Survival/drug effects , Humans , Liver/drug effects , Liver Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...