Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Front Immunol ; 14: 1157263, 2023.
Article in English | MEDLINE | ID: mdl-37081876

ABSTRACT

Introduction: The rapid development of vaccines to prevent COVID-19 has raised the need to compare the capacity of different vaccines in terms of developing a protective humoral response. Previous studies have shown inconsistent results in this area, highlighting the importance of further research to evaluate the efficacy of different vaccines. Methods: This study utilized a highly sensitive and reliable flow cytometry method to measure the titers of IgG1 isotype antibodies in the blood of healthy volunteers after receiving one or two doses of various vaccines administered in Spain. The method was also used to simultaneously measure the reactivity of antibodies to the S protein of the original Wuhan strain and variants B.1.1.7 (Alpha), B.1.617.2 (Delta), and B.1.617.1 (Kappa). Results: Significant differences were observed in the titer of anti-S antibodies produced after a first dose of the vaccines ChAdOx1 nCov-19/AstraZeneca, mRNA-1273/Moderna, BNT162b2/Pfizer-BioNTech, and Ad26.COV.S/Janssen. Furthermore, a relative reduction in the reactivity of the sera with the Alpha, Delta, and Kappa variants, compared to the Wuhan strain, was observed after the second boosting immunization. Discussion: The findings of this study provide a comparison of different vaccines in terms of anti-S antibody generation and cast doubts on the convenience of repeated immunization with the same S protein sequence. The multiplexed capacity of the flow cytometry method utilized in this study allowed for a comprehensive evaluation of the efficacy of various vaccines in generating a protective humoral response. Future research could focus on the implications of these findings for the development of effective COVID-19 vaccination strategies.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Antibody Formation , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , ChAdOx1 nCoV-19 , Spike Glycoprotein, Coronavirus , Vaccination , Antibodies
2.
Front Immunol ; 13: 809285, 2022.
Article in English | MEDLINE | ID: mdl-35296086

ABSTRACT

The emergence of COVID-19 has led to a worldwide challenge for the rapid development of vaccines. Several types of safe and effective vaccines have been available in a time frame never seen before. Now that several hundred million people have been vaccinated there is an opportunity to compare vaccines in terms of protection and immune response. Here, we have applied a highly sensitive multiplexed flow cytometry method to measure simultaneously IgM, IgG1 and IgA anti-spike protein antibodies generated in response to three vaccines: ChAdOx1 (Oxford-AstraZeneca), mRNA-1273 (Moderna), and BNT162b2 (Pfizer-BioNTech). We have found that mRNA vaccines (mRNA-1273 and BNT162b2) induce a stronger humoral response, both after the first and the second dose, than the adenovirus-based ChAdOx1 vaccine. We also found that, in the elderly, antibody titers negatively correlate with the age of the donor but, also, that antibody titers remain stable for at least 6 months after complete vaccination. Finally, we found that one dose of BNT162b2 is sufficient to induce the highest antibody titers in seropositive pre-vaccination donors. We hope these data will help to guide future decisions on vaccination strategies.


Subject(s)
COVID-19 , Vaccines , Aged , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Infant , SARS-CoV-2
3.
Oncotarget ; 5(22): 11308-18, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25350956

ABSTRACT

Triple negative breast cancers (TNBCs) account for 15% of all breast cancers, and represent one of the most aggressive forms of the disease, exhibiting short relapse-free survival. In contrast to other breast cancer subtypes, the absence of knowledge about the etiopathogenic alterations that cause TNBCs force the use of chemotherapeutics to treat these tumors. Because of this, efforts have been devoted with the aim of incorporating novel therapies into the clinical setting. Kinases play important roles in the pathophysiology of several tumors, including TNBC. Since expression of the MAP kinase ERK5 has been linked to patient outcome in breast cancer, we analyzed the potential value of its targeting in TNBC. ERK5 was frequently overexpressed and active in samples from patients with TNBC, as well as in explants from mice carrying genetically-defined TNBC tumors. Moreover, expression of ERK5 was linked to a worse prognosis in TNBC patients. Knockdown experiments demonstrated that ERK5 supported proliferation of TNBC cells. Pharmacological inhibition of ERK5 with TG02, a clinical stage inhibitor which targets ERK5 and other kinases, inhibited cell proliferation by blocking passage of cells through G1 and G2, and also triggered apoptosis in certain TNBC cell lines. TG02 had significant antitumor activity in a TNBC xenograft model in vivo, and also augmented the activity of chemotherapeutic agents commonly used to treat TNBC. Together, these data indicate that ERK5 targeting may represent a valid strategy against TNBC, and support the development of trials aimed at evaluating the clinical effectiveness of drugs that block this kinase.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/enzymology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Docetaxel , Female , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Humans , Mice , Mitogen-Activated Protein Kinase 7/metabolism , Molecular Targeted Therapy , Random Allocation , Taxoids/administration & dosage , Taxoids/pharmacology , Triple Negative Breast Neoplasms/pathology , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinblastine/pharmacology , Vinorelbine , Xenograft Model Antitumor Assays
4.
Clin Cancer Res ; 19(10): 2677-87, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23532886

ABSTRACT

PURPOSE: To analyze the antimyeloma potential of TG02, an ERK5/CDK inhibitory drug. EXPERIMENTAL DESIGN: Utilizing different multiple myeloma cell lines we determined the effect of TG02 over viability by MTT assays. The apoptotic effect over multiple myeloma patient samples was studied ex vivo by cytometry. The mechanism of action of TG02 was analyzed in the cell line MM1S, studying its effect on the cell cycle, the induction of apoptosis, and the loss of mitochondrial membrane potential by cytometry and Western blot. Two models of multiple myeloma xenograft were utilized to study the in vivo action of TG02. RESULTS: TG02 potently inhibited proliferation and survival of multiple myeloma cell lines, even under protective bone marrow niche conditions, and selectively induced apoptosis of primary patient-derived malignant plasma cells. TG02 displayed significant single-agent activity in two multiple myeloma xenograft models, and enhanced the in vivo activity of bortezomib and lenalidomide. Signaling analyses revealed that the drug simultaneously blocked the activity of CDKs 1, 2, and 9 as well as the MAP kinase ERK5 in MM1S cells, leading to cell-cycle arrest and rapid commitment to apoptosis. TG02 induced robust activation of both the intrinsic and extrinsic pathways of apoptosis, and depletion of XIAP and the key multiple myeloma survival protein Mcl-1. CONCLUSIONS: TG02 is a promising new antimyeloma agent that is currently in phase I clinical trials in leukemia and multiple myeloma patients.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Multiple Myeloma/prevention & control , Xenograft Model Antitumor Assays , Animals , Blotting, Western , Boronic Acids/pharmacology , Bortezomib , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lenalidomide , Mice, SCID , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Cyclin-Dependent Kinase-Activating Kinase
5.
Haematologica ; 97(8): 1218-24, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22371183

ABSTRACT

UNLABELLED: Background Recent findings suggest that a specific deletion of Dicer1 in mesenchymal stromal cell-derived osteoprogenitors triggers several features of myelodysplastic syndrome in a murine model. Our aim was to analyze DICER1 and DROSHA gene and protein expression in mesenchymal stromal cells (the osteoblastic progenitors) obtained from bone marrow of myelodysplastic syndrome patients, in addition to microRNA expression profile and other target genes such as SBDS, a DICER1-related gene that promotes bone marrow dysfunction and myelodysplasia when repressed in a murine model. DESIGN AND METHODS: Mesenchymal stromal cells from 33 bone marrow samples were evaluated. DICER, DROSHA and SBDS gene expression levels were assessed by real-time PCR and protein expression by Western blot. MicroRNA expresion profile was analyzed by commercial low-density arrays and some of these results were confirmed by individual real-time PCR. RESULTS: Mesenchymal stromal cells from myelodysplastic syndrome patients showed lower DICER1 (0.65±0.08 vs. 1.91±0.57; P=0.011) and DROSHA (0.62±0.06 vs. 1.38±0.29; P=0.009) gene expression levels, two relevant endonucleases associated to microRNA biogenesis, in comparison to normal myelodysplastic syndrome. These findings were confirmed at protein levels by Western blot. Strikingly, no differences were observed between paired mononuclear cells from myelodysplastic syndrome and controls. In addition, mesenchymal stromal cells from myelodysplastic syndrome patients showed significant lower SBDS (0.63±0.06 vs. 1.15±0.28; P=0.021) gene expression levels than mesenchymal stromal cells from healthy controls. Furthermore, mesenchymal stromal cells from myelodysplastic syndrome patients showed an underlying microRNA repression compared to healthy controls. Real-time PCR approach confirmed that mir-155, miR-181a and miR-222 were down-expressed in mesenchymal stromal cells from myelodysplastic syndrome patients. Conclusions This is the first description of an impaired microRNA biogenesis in human mesenchymal stromal cells from myelodysplastic syndrome patients, where DICER1 and DROSHA gene and protein downregulation correlated to a gene and microRNA abnormal expression profile, validating the animal model results previously described.


Subject(s)
DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Neoplastic , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Myelodysplastic Syndromes/genetics , Proteins/genetics , Ribonuclease III/genetics , Aged , Aged, 80 and over , Case-Control Studies , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Myelodysplastic Syndromes/metabolism
6.
PLoS One ; 6(11): e27583, 2011.
Article in English | MEDLINE | ID: mdl-22132115

ABSTRACT

Multiple myeloma (MM) is a hematological disease characterized by an abnormal accumulation of plasma cells in the bone marrow. These cells have frequent cytogenetic abnormalities including translocations of the immunoglobulin heavy chain gene and chromosomal gains and losses. In fact, a singular characteristic differentiating MM from other hematological malignancies is the presence of a high degree of aneuploidies. As chromosomal abnormalities can be generated by alterations in the spindle assembly checkpoint (SAC), the functionality of such checkpoint was tested in MM. When SAC components were analyzed in MM cell lines, the RNA levels of most of them were conserved. Nevertheless, the protein content of some key constituents was very low in several cell lines, as was the case of MAD2 or CDC20 in RPMI-8226 or RPMI-LR5 cells. The recovery of their cellular content did not substantially affect cell growth, but improved their ability to segregate chromosomes. Finally, SAC functionality was tested by challenging cells with agents disrupting microtubule dynamics. Most of the cell lines analyzed exhibited functional defects in this checkpoint. Based on the data obtained, alterations both in SAC components and their functionality have been detected in MM, pointing to this pathway as a potential target in MM treatment.


Subject(s)
M Phase Cell Cycle Checkpoints , Multiple Myeloma/pathology , Aneuploidy , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Genomic Instability/drug effects , Humans , M Phase Cell Cycle Checkpoints/genetics , Mad2 Proteins , Multiple Myeloma/genetics , Nocodazole/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Retroviridae/drug effects , Retroviridae/genetics , Transduction, Genetic
7.
Haematologica ; 96(5): 687-95, 2011 May.
Article in English | MEDLINE | ID: mdl-21330323

ABSTRACT

BACKGROUND: Although the majority of patients with acute myeloid leukemia initially respond to conventional chemotherapy, relapse is still the leading cause of death, probably because of the presence of leukemic stem cells that are insensitive to current therapies. We investigated the antileukemic activity and mechanism of action of zalypsis, a novel alkaloid of marine origin. DESIGN AND METHODS: The activity of zalypsis was studied in four acute myeloid leukemia cell lines and in freshly isolated blasts taken from patients with acute myeloid leukemia before they started therapy. Zalypsis-induced apoptosis of both malignant and normal cells was measured using flow cytometry techniques. Gene expression profiling and western blot studies were performed to assess the mechanism of action of the alkaloid. RESULTS: Zalypsis showed a very potent antileukemic activity in all the cell lines tested and potentiated the effect of conventional antileukemic drugs such as cytarabine, fludarabine and daunorubicin. Interestingly, zalypsis showed remarkable ex vivo potency, including activity against the most immature blast cells (CD34(+) CD38(-) Lin(-)) which include leukemic stem cells. Zalypsis-induced apoptosis was the result of an important deregulation of genes involved in the recognition of double-strand DNA breaks, such as Fanconi anemia genes and BRCA1, but also genes implicated in the repair of double-strand DNA breaks, such as RAD51 and RAD54. These gene findings were confirmed by an increase in several proteins involved in the pathway (pCHK1, pCHK2 and pH2AX). CONCLUSIONS: The potent and selective antileukemic effect of zalypsis on DNA damage response mechanisms observed in acute myeloid leukemia cell lines and in patients' samples provides the rationale for the investigation of this compound in clinical trials.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA Damage , Stem Cells/drug effects , Tetrahydroisoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Oligonucleotide Array Sequence Analysis , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Stem Cells/pathology , Tumor Cells, Cultured
8.
Blood ; 113(16): 3781-91, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19020308

ABSTRACT

Multiple myeloma (MM) remains incurable, and new drugs with novel mechanisms of action are still needed. In this report, we have analyzed the action of Zalypsis, an alkaloid analogous to certain natural marine compounds, in MM. Zalypsis turned out to be the most potent antimyeloma agent we have tested so far, with IC(50) values from picomolar to low nanomolar ranges. It also showed remarkable ex vivo potency in plasma cells from patients and in MM cells in vivo xenografted in mice. Besides the induction of apoptosis and cell cycle arrest, Zalypsis provoked DNA double-strand breaks (DSBs), evidenced by an increase in phospho-histone-H2AX and phospho-CHK2, followed by a striking overexpression of p53 in p53 wild-type cell lines. In addition, in those cell lines in which p53 was mutated, Zalypsis also provoked DSBs and induced cell death, although higher concentrations were required. Immunohistochemical studies in tumors also demonstrated histone-H2AX phosphorylation and p53 overexpression. Gene expression profile studies were concordant with these results, revealing an important deregulation of genes involved in DNA damage response. The potent in vitro and in vivo antimyeloma activity of Zalypsis uncovers the high sensitivity of tumor plasma cells to DSBs and strongly supports the use of this compound in MM patients.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Breaks, Double-Stranded/drug effects , Multiple Myeloma/drug therapy , Tetrahydroisoquinolines/pharmacology , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/therapeutic use , Cell Death , Checkpoint Kinase 2 , Dose-Response Relationship, Drug , Histones/genetics , Histones/metabolism , Humans , Mice , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Mutation , Phosphorylation/drug effects , Plasma Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tetrahydroisoquinolines/therapeutic use , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
9.
Haematologica ; 93(1): 57-66, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18166786

ABSTRACT

BACKGROUND: Proteasome inhibition represents a promising novel anticancer therapy, and bortezomib is a highly selective reversible inhibitor of the proteasome complex. Acute myeloid leukemia (AML) is an immnunophenotypically heterogeneous group of diseases, with CD34(+) cases being associated with drug resistance and poor outcome. We investigated the effects of bortezomib on the growth and survival of AML cells. DESIGN AND METHODS: We studied the in vitro activity and mechanism of action of bortezomib on both cell lines and fresh cells from 28 AML patients including CD34(+) and CD34(-) cases. RESULTS: Bortezomib showed potent anti-AML activity (IC(50) < 50 nM), which was greater than that of conventional agents (doxorubicin, cytarabine and fludarabine). Moreover, synergistic effects were observed when bortezomib was administered in combination with doxorubicin and cytarabine. Mechanistically, bortezomib induced accumulation of cells in the G(2)/M phase, with up-regulation of p27, together with cell death through an increase in the mitochondrial outer membrane permeability involving caspase-dependent and -independent pathways. The apoptotic activity of bortezomib on fresh CD34(+) blast cells from patients was similar to that observed on CD34(-)blast cells. Importantly, bortezomib was significantly more active than doxorubicin in the immature CD34(+) cells, while there were no differences in its action on CD34(-) cells. CONCLUSIONS: Bortezomib induces apoptosis in acute myeloid leukemia cells in vitro. Whether this drug might be useful in the treatment of patients with acute myeloid leukemia can be established only in ad hoc clinical trials.


Subject(s)
Antigens, CD34/biosynthesis , Boronic Acids/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Aged , Antineoplastic Agents/pharmacology , Apoptosis , Bortezomib , Cell Line, Tumor , Humans , Inhibitory Concentration 50 , Leukemia, Myeloid, Acute/metabolism , Middle Aged , Phenotype , Proteasome Endopeptidase Complex/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...