Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Curr Med Chem ; 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39225211

RESUMO

BACKGROUND: Selective Cyclin-Dependent Kinase 4/6 inhibitors (CDK4/6i) have revolutionized the treatment of breast cancer and have potential in other cancers, being manageable drugs yet with some bone marrow toxicity. Selective CDK9 inhibitors (CDK9i) never advanced into clinical use, partly due to side effects, including gastrointestinal toxicity, and a small window between activity and cytotoxicity, which results in a narrow therapeutic index (TI). METHOD: To overcome the drawbacks of CDK4/6 and CDK9 inhibitors, we have developed myrtleciclib, a selective CDK4/6/9 inhibitor with few non-critical molecular off-targets. RESULTS: Myrtleciclib appears to bind to an allosteric site, unlike all other CDK4/6i and CDK9i acting by an ATP-competitive mechanism, which supports target specificity. Myrtleciclib's anti-proliferative effects are greater and its Therapeutic Index (TI) is broader than CDK9 and CDK4/6-only inhibitors. This can be explained by a moderate target inhibition, resulting in limited cytotoxicity. Moreover, we documented a synergy between CDK9 and CDK4/6 pathways inhibition, justifying increased drug efficacy, yet such synergy can only be achieved when the inhibition of both CDK9 and CDK4/6 is embedded within the same molecule and balanced within a certain ratio, as it is the case with myrtleciclib. Unlike CDK4/6i, myrtleciclib also induces cell death and apoptosis selectively on cancer cell lines, not on bystander cells. Synergy between myrtleciclib and other drugs with complementary Mechanism of Action (MoA) has also been documented. CONCLUSION: CDK4/6/9i might represent a new frontier in cancer treatment to overcome the limitations of CDK4/6i and CDK9i for the treatment of cancers, including aggressive cancers with high unmet needs.

2.
Viruses ; 15(10)2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37896888

RESUMO

SARS-CoV-2 is inactivated in aerosol (its primary mode of transmission) by means of radiated microwaves at frequencies that have been experimentally determined. Such frequencies are best predicted by the mathematical model suggested by Taylor, Margueritat and Saviot. The alignment between such mathematical prediction and the outcomes of our experiments serves to reinforce the efficacy of the radiated microwave technology and its promise in mitigating the transmission of SARS-CoV-2 in its naturally airborne state.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Micro-Ondas , Aerossóis e Gotículas Respiratórios , Modelos Teóricos
3.
Viruses ; 15(7)2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37515131

RESUMO

Coronaviruses are a family of viruses that cause disease in mammals and birds. In humans, coronaviruses cause infections on the respiratory tract that can be fatal. These viruses can cause both mild illnesses such as the common cold and lethal illnesses such as SARS, MERS, and COVID-19. Air transmission represents the principal mode by which people become infected by SARS-CoV-2. To reduce the risks of air transmission of this powerful pathogen, we devised a method of inactivation based on the propagation of electromagnetic waves in the area to be sanitized. We optimized the conditions in a controlled laboratory environment mimicking a natural airborne virus transmission and consistently achieved a 90% (tenfold) reduction of infectivity after a short treatment using a Radio Frequency (RF) wave emission with a power level that is safe for people according to most regulatory agencies, including those in Europe, USA, and Japan. To the best of our knowledge, this is the first time that SARS-CoV-2 has been shown to be inactivated through RF wave emission under conditions compatible with the presence of human beings and animals. Additional in-depth studies are warranted to extend the results to other viruses and to explore the potential implementation of this technology in different environmental conditions.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Humanos , Micro-Ondas , Aerossóis e Gotículas Respiratórios , Europa (Continente) , Mamíferos
4.
Nanomaterials (Basel) ; 13(24)2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38132987

RESUMO

The emergence of SARS-CoV-2 variants requires close monitoring to prevent the reoccurrence of a new pandemic in the near future. The Omicron variant, in particular, is one of the fastest-spreading viruses, showing a high ability to infect people and evade neutralization by antibodies elicited upon infection or vaccination. Therefore, the search for broad-spectrum antivirals that can inhibit the infectious capacity of SARS-CoV-2 is still the focus of intense research. In the present work, hyperbranched poly-L-lysine nanopolymers, which have shown an excellent ability to block the original strain of SARS-CoV-2 infection, were modified with L-arginine. A thermal reaction at 240 °C catalyzed by boric acid yielded Lys-Arg hyperbranched nanopolymers. The ability of these nanopolymers to inhibit viral replication were assessed for the original, Delta, and Omicron strains of SARS-CoV-2 together with their cytotoxicity. A reliable indication of the safety profile and effectiveness of the various polymeric compositions in inhibiting or suppressing viral infection was obtained by the evaluation of the therapeutic index in an in vitro prevention model. The hyperbranched L-arginine-modified nanopolymers exhibited a twelve-fold greater therapeutic index when tested with the original strain. The nanopolymers could also effectively limit the replication of the Omicron strain in a cell culture.

5.
PLoS One ; 17(11): e0276751, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36355808

RESUMO

Despite new antivirals are being approved against SARS-CoV-2 they suffer from significant constraints and are not indicated for hospitalized patients, who are left with few antiviral options. Repurposed drugs have previously shown controversial clinical results and it remains difficult to understand why certain trials delivered positive results and other trials failed. Our manuscript contributes to explaining the puzzle: this might have been caused by a suboptimal drug exposure and, consequently, an incomplete virus suppression, also because the drugs have mostly been used as add-on monotherapies. As with other viruses (e.g., HIV and HCV) identifying synergistic combinations among such drugs could overcome monotherapy-related limitations. In a cell culture model for SARS-CoV-2 infection the following stringent criteria were adopted to assess drug combinations: 1) identify robust, synergistic antiviral activity with no increase in cytotoxicity, 2) identify the lowest drug concentration inhibiting the virus by 100% (LIC100) and 3) understand whether the LIC100 could be reached in the lung at clinically indicated drug doses. Among several combinations tested, remdesivir with either azithromycin or ivermectin synergistically increased the antiviral activity with no increase in cytotoxicity, improving the therapeutic index and lowering the LIC100 of every one of the drugs to levels that are expected to be achievable and maintained in the lung for a therapeutically relevant period of time. These results are consistent with recent clinical observations showing that intensive care unit admission was significantly delayed by the combination of AZI and RDV, but not by RDV alone, and could have immediate implications for the treatment of hospitalized patients with COVID-19 as the proposed "drug cocktails" should have antiviral activity against present and future SARS-CoV-2 variants without significant overlapping toxicity, while minimizing the onset of drug resistance. Our results also provide a validated methodology to help sort out which combination of drugs are most likely to be efficacious in vivo, based on their in vitro activity, potential synergy and PK profiles.


Assuntos
Tratamento Farmacológico da COVID-19 , SARS-CoV-2 , Humanos , Antivirais/farmacologia , Antivirais/uso terapêutico , Monofosfato de Adenosina/farmacologia , Monofosfato de Adenosina/uso terapêutico , Alanina/farmacologia , Alanina/uso terapêutico , Pulmão , Combinação de Medicamentos
6.
Nanoscale ; 13(39): 16465-16476, 2021 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-34553728

RESUMO

The coronavirus pandemic (COVID-19) had spread rapidly since December 2019, when it was first identified in Wuhan, China. As of April 2021, more than 130 million cases have been confirmed, with more than 3 million deaths, making it one of the deadliest pandemics in history. Different approaches must be put in place to confront a new pandemic: community-based behaviours (i.e., isolation and social distancing), antiviral treatments, and vaccines. Although behaviour-based actions have produced significant benefits and several efficacious vaccines are now available, there is still an urgent need for treatment options. Remdesivir represents the first antiviral drug approved by the Food and Drug Administration for COVID-19 but has several limitations in terms of safety and treatment benefits. There is still a strong request for other effective, safe, and broad-spectrum antiviral systems in light of future emergent coronaviruses. Here, we describe a polymeric nanomaterial derived from L-lysine, with an antiviral activity against SARS-CoV-2 associated with a good safety profile in vitro. Nanoparticles of hyperbranched polylysine, synthesized by L-lysine's thermal polymerization catalyzed by boric acid, effectively inhibit the SARS-CoV-2 replication. The virucidal activity is associated with the charge and dimension of the nanomaterial, favouring the electrostatic interaction with the viral surface being only slightly larger than the virions' dimensions. Low-cost production and easiness of synthesis strongly support the further development of such innovative nanomaterials as a tool for potential treatments of COVID-19 and, in general, as broad-spectrum antivirals.


Assuntos
Antivirais , COVID-19 , Antivirais/farmacologia , Humanos , Pandemias , Polilisina , SARS-CoV-2
7.
ACS Chem Biol ; 13(1): 253-266, 2018 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-29235845

RESUMO

HIV/AIDS is still one of the leading causes of death worldwide. Current drugs that target the canonical steps of the HIV-1 life cycle are efficient in blocking viral replication but are unable to eradicate HIV-1 from infected patients. Moreover, drug resistance (DR) is often associated with the clinical use of these molecules, thus raising the need for novel drug candidates as well as novel putative drug targets. In this respect, pharmacological inhibition of the highly conserved and multifunctional nucleocapsid protein (NC) of HIV-1 is considered a promising alternative to current drugs, particularly to overcome DR. Here, using a multidisciplinary approach combining in silico screening, fluorescence-based molecular assays, and cellular antiviral assays, we identified nordihydroguaiaretic acid (6), as a novel natural product inhibitor of NC. By using NMR, mass spectrometry, fluorescence spectroscopy, and molecular modeling, 6 was found to act through a dual mechanism of action never highlighted before for NC inhibitors (NCIs). First, the molecule recognizes and binds NC noncovalently, which results in the inhibition of the nucleic acid chaperone properties of NC. In a second step, chemical oxidation of 6 induces a potent chemical inactivation of the protein. Overall, 6 inhibits NC and the replication of wild-type and drug-resistant HIV-1 strains in the low micromolar range with moderate cytotoxicity that makes it a profitable tool compound as well as a good starting point for the development of pharmacologically relevant NCIs.


Assuntos
Fármacos Anti-HIV/química , Fármacos Anti-HIV/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , HIV-1/efeitos dos fármacos , Proteínas do Nucleocapsídeo/antagonistas & inibidores , Fármacos Anti-HIV/toxicidade , Apoptose/efeitos dos fármacos , Farmacorresistência Viral/efeitos dos fármacos , HIV-1/fisiologia , Humanos , Concentração Inibidora 50 , Leucócitos Mononucleares/efeitos dos fármacos , Espectroscopia de Ressonância Magnética , Mitocôndrias/efeitos dos fármacos , Modelos Moleculares , Proteínas do Nucleocapsídeo/química , Espectrometria de Fluorescência , Relação Estrutura-Atividade , Replicação Viral/efeitos dos fármacos
8.
Curr Drug Deliv ; 3(1): 83-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16472097

RESUMO

DermaVir employs a topical, non-invasive method for vaccine delivery to dendritic cells. The vaccine product contains plasmid DNA as the active ingredient, encoding authentically expressed retroviral genes with appropriate safety modifications. The non-viral delivery system packages the DNA within pathogen-like nanoparticles and studies indicate that vaccine antigens are taken up by epidermal Langerhans cells, the precursors of dendritic cells. DermaVir loaded dendritic cells reach the draining lymph node target but not the bloodstream nor indiscriminately other organ systems. Safety data from DermaVir immunized infected macaques indicate improved survival, absence of apparent toxicities other than transient erythema and lack of recombination between the vaccine DNA and the infectious viral DNA integrated in the host genome. DermaVir represents a potential new approach for the treatment of HIV infection to be utilized either in conjunction with antiretroviral therapy or during structured treatment interruption.


Assuntos
Vacinas contra a AIDS/administração & dosagem , Células Dendríticas/imunologia , Infecções por HIV/terapia , Administração Cutânea , Animais , Diferenciação Celular , DNA/genética , DNA/metabolismo , Antígenos HIV/imunologia , Infecções por HIV/imunologia , Células de Langerhans/citologia , Células de Langerhans/imunologia , Linfonodos/citologia , Linfonodos/imunologia , Macaca mulatta , Camundongos , Plasmídeos/genética , Vírus da Imunodeficiência Símia/imunologia , Baço/metabolismo
9.
Chin Med J (Engl) ; 119(19): 1629-38, 2006 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-17042976

RESUMO

BACKGROUND: The mechanism of chronic immune activation and impairment of HIV-specific immune responses during chronic infection is not fully understood. However, it is known that high immune activation leads to more rapid progression to AIDS. We hypothesize that CD4(+) T cell-mediated viral antigen presentation contributes to this pathologic immune activation in HIV-infected individuals. METHODS: HIV-specific T cells, responding to noninfectious HIV-1 virions as antigen, were measured by flow cytometric assays. These experimental conditions reflect the in vivo condition where noninfectious HIV-1 represents more than 99% of the antigens. RESULTS: CD4(+) T cells purified from HIV-infected individuals were capable of cross presenting exogenous noninfectious HIV-1 virions to HIV-1-specific CD8(+) T cells. Cross presentation required the entry of HIV-1 to CD4(+) T cells and antigen translocation from endoplasmic reticulum to the Golgi complex. Blocking CD4(+) mediated activation of HIV-specific CD8(+) T cells and redirecting the viral antigens to antigen presenting cells improved HIV-specific T cell responses. CONCLUSIONS: One possible cause of chronic immune activation and impairment of HIV-1 specific T cell responses is represented by HIV-1 harboring CD4(+) T cells cross presenting HIV-1 antigen to activate CD8(+) T cells. This new mechanism provides the first evidence that cross presentation of noninfectious HIV-1 virions play a role in the immunopathogenesis of HIV-1 infection.


Assuntos
Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , HIV-1/imunologia , Vírion/imunologia , Adulto , Linfócitos T CD4-Positivos/virologia , Humanos , Ativação Linfocitária , Masculino
10.
AIDS ; 19(11): 1173-81, 2005 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-15990570

RESUMO

BACKGROUND: The hydroxyurea-didanosine combination has been shown to limit immune activation (a major pathogenic component of HIV/AIDS) and suppress viral load by both antiviral and cytostatic ('virostatic') activities. Virostatics action represent a novel approach to attack HIV/AIDS from multiple directions; however, the use of these drugs is limited by the lack of understanding of their dose-dependent mechanism of action and by fear of pancreatic toxicity, even though a large review of ACTG studies has shown that hydroxyurea does not increase the incidence of pancreatitis. METHODS: In vitro cytostatic and cytotoxic activity, inhibition of viral replication and immune activation by pharmacologically attainable plasma concentrations of hydroxyurea (10-100 micromol/l) and didanosine (1-5 micromol/l) were analyzed by cell proliferation, viability, apoptosis and infection assays using peripheral blood mononuclear cells. In vivo, 600, 900 and 1200 mg daily doses of hydroxyurea in combination with standard doses of didanosine and stavudine were studied in 115 randomized chronically infected patients. RESULTS: A cytostatic low (10 micromol/l) concentration of hydroxyurea inhibited cell proliferation and HIV replication in vitro. A gradual switch from cytostatic to cytotoxic effects was observed by increasing hydroxyurea concentration to 50-100 micromol/l, predicting that lower doses of hydroxyurea would be less toxic and more potent in vivo. The clinical results confirmed that 600 mg hydroxyurea was better tolerated, had fewer side effects and was more potent in suppressing HIV replication than the higher doses. CONCLUSIONS: A bimodal, dose-dependent, cytostatic-cytotoxic switch is an immune-based mechanism explaining the apparent paradox that lowering the dose of hydroxyurea to 600 mg daily induces maximal antiviral suppression in HIV-infected patients.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Didanosina/administração & dosagem , Infecções por HIV/tratamento farmacológico , Hidroxiureia/administração & dosagem , Inibidores da Síntese de Ácido Nucleico/administração & dosagem , Replicação Viral/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Humanos , Hidroxiureia/efeitos adversos , Dose Máxima Tolerável , Inibidores da Síntese de Ácido Nucleico/efeitos adversos , Pancreatopatias/induzido quimicamente , Carga Viral
11.
AIDS ; 19(2): 137-44, 2005 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-15668538

RESUMO

OBJECTIVE: To demonstrate that, despite a dose-dependent cytostatic effect, hydroxyurea (HU) does not have immunosuppressive effects. METHODS: The effects of HU on T lymphocyte proliferation parameters, activation phenotype and cytokine production were examined in vitro after exposure to clinically relevant concentrations of HU (10, 50, and 100 micromol/l). The effects of HU in vivo on CD4 T cell counts, viral load, activation phenotype and virus-specific response were examined in 17 Rhesus macaques infected with SIV(mac251) and randomized into three groups: untreated controls; treated with (R)-9-(2-phosphonylmethoxypropyl)adenine (PMPA) and didanosine (ddI) only; and treated with PMPA, didanosine, and HU. RESULTS: The in vitro inhibition of T lymphocyte proliferation confirmed the cytostatic effect of HU, with a linear dose-dependent effect; however, no relevant differences were found in the expression of activation markers between treated and untreated controls. Both T helper type 1 and type 2 cytokine production were enhanced by HU. Consistent with the in vitro results, a blunted increase of peripheral CD4 T cells was observed in vivo in the HU group, without relevant effects on the expression of activation markers, and SIV-specific T cell responses were not affected by HU. CONCLUSIONS: Hyper-proliferation of T-lymphocytes is a major factor contributing to HIV pathogenesis. HU exerts a cytostatic effect on T lymphocytes, without altering their activation and apparently without having an immunosuppressive effect. The increase in cytokine production at the single cell level might compensate for the decrease in the percentage of activated CD4 T lymphocytes, without overall impairment of HIV-specific immune responses.


Assuntos
Antineoplásicos/farmacologia , Hidroxiureia/farmacologia , Imunossupressores/farmacologia , Linfócitos T/efeitos dos fármacos , Animais , Antígenos CD/imunologia , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/imunologia , Relação Dose-Resposta a Droga , Humanos , Interferon gama/imunologia , Ativação Linfocitária/efeitos dos fármacos , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia
12.
AIDS ; 19(1): 35-43, 2005 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-15627031

RESUMO

OBJECTIVE: To reconstitute immune responses capable of eliminating infected cells and suppressing viral load during chronic retroviral infection. DESIGN: : A topical, DNA-based therapeutic immunization (DermaVir) was designed to express most of the regulatory and structural viral genes in dendritic cells. METHODS: DermaVir alone and in combination with antiretroviral drugs was tested in chronically SIV-infected macaques. RESULTS: DermaVir provided virological, immunological and clinical benefit for SIV-infected macaques during chronic infection and AIDS. In combination with antiretroviral drugs, DermaVir augmented SIV-specific T-cell responses and enhanced control of viral load rebound during treatment interruptions. CONCLUSIONS: The results indicate the feasibility of therapeutic immunization even in immune compromised hosts, and suggest that DermaVir can complement antiretroviral drugs to sustain suppression of HIV-1 replication.


Assuntos
Vacinas contra a AIDS/uso terapêutico , Terapia Antirretroviral de Alta Atividade/métodos , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Vacinas contra a AIDS/efeitos adversos , Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Síndrome da Imunodeficiência Adquirida/imunologia , Síndrome da Imunodeficiência Adquirida/virologia , Animais , Linfócitos T CD4-Positivos/imunologia , Doença Crônica , Estudos de Coortes , Células Dendríticas/imunologia , Quimioterapia Combinada , Feminino , Imunização/métodos , Contagem de Linfócitos , Macaca , Masculino , RNA Viral/análise , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Suínos , Carga Viral/métodos , Replicação Viral/imunologia
13.
J Invest Dermatol ; 124(1): 160-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15654970

RESUMO

Human immunodeficiency virus (HIV) vaccines have the potential to improve antiretroviral drug treatment by inducing cytotoxic killing of HIV-infected cells. Prophylactic vaccines utilize new antigens to initiate immunity; however, in HIV-infected individuals the load of viral antigen is not the limiting factor for the restoration of immune responses. Here we describe a novel immunization strategy with DermaVir that improves viral antigen presentation using dendritic cells (DC). DermaVir contains a distinctive plasmid DNA expressing all HIV proteins except integrase to induce immune responses with broad specificity. The DNA is formulated to a mannosilated particle to target antigen-presenting cells and to protect the DNA from intracellular degradation. After topical application, DermaVir-transduced cells migrate from the skin to the draining lymph node and interdigitate as DermaVir-expressing, antigen-presenting DC. We compared the immunogenicity of topical and ex vivo DC-based DermaVir vaccinations in naive rhesus macaques. Both vaccinations induced simian immunodeficiency virus-specific CD4 helper and CD8 memory T cells detected by an in vivo skin test and an in vitro intracellular cytokine-based assay. Topical DermaVir vaccination represents an improvement upon existing ex vivo DC-based immunization technologies and may provide a new therapeutic option for HIV-infected patients.


Assuntos
Vacinas contra a AIDS/imunologia , Vacinas contra a AIDS/farmacologia , Síndrome da Imunodeficiência Adquirida/prevenção & controle , Vacinas contra a AIDS/genética , Síndrome da Imunodeficiência Adquirida/imunologia , Administração Tópica , Animais , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Feminino , Regulação Viral da Expressão Gênica/imunologia , Linfonodos/citologia , Linfonodos/imunologia , Macaca mulatta , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos , Células Th1/imunologia , Vacinação/métodos
14.
AIDS Res Hum Retroviruses ; 21(4): 263-72, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15943568

RESUMO

The goal of this study was to optimize the hydroxyurea dosage in HIV-infected patients, and to minimize the toxicity and maximize the antiviral efficacy of the hydroxyurea-didanosine combination. In a randomized, open-label study (RIGHT 702, a multicenter trial performed in private and institutional practices), three daily doses (600 microg, 800-900 microg, and 1200 microg) of hydroxyurea were administered in combination with didanosine and stavudine to 115 chronically HIV-infected patients, one-third antiretroviral drug naive, with viremia between 5000 and 200,000 copies/ml regardless of CD4+ cell count. The primary efficacy end point was the proportion of patients with plasma HIV-1 RNA levels below 400 copies/ml after 24 weeks of therapy. In the RIGHT 702 intent-to-treat population the lowest (600 mg) dose of hydroxyurea was better tolerated, associated with fewer adverse events, and more potent by all efficacy parameters, including the primary end point (76 versus 60% patients with viremia<400 copies/ml at week 24 for the 600-mg and 800- to 900-mg dose groups, respectively; p=0.027), the mean area under the curve (60.3 versus 65.8; p=0.016), and the mean log10 decrease (-1.95 versus -0.77; p=0.001). Patients receiving 600 mg of hydroxyurea daily also had the highest CD4+ cell count, CD4+/CD8+ cell ratio, and lowest CD8+ cell count and percentage (p=0.035). The RIGHT 702 trial provides an explanation for the increased toxicity and decreased efficacy of hydroxyurea when it was used at high dosage (1200 mg daily). At the optimal dosage of 600 mg daily, hydroxyurea, in combination with didanosine, deserves reevaluation for the long-term management of HIV/AIDS worldwide, because of its excellent resistance profile, durability, and affordability.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Didanosina/administração & dosagem , Infecções por HIV/tratamento farmacológico , Hidroxiureia/administração & dosagem , Hidroxiureia/efeitos adversos , Fármacos Anti-HIV/efeitos adversos , Fármacos Anti-HIV/uso terapêutico , Contagem de Linfócito CD4 , Relação CD4-CD8 , Didanosina/uso terapêutico , Quimioterapia Combinada , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Feminino , HIV , Infecções por HIV/virologia , Humanos , Hidroxiureia/uso terapêutico , Masculino , RNA Viral/sangue , Estavudina/administração & dosagem , Estavudina/uso terapêutico , Carga Viral , Viremia
15.
Blood ; 112(4): 930-1, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18684877
16.
AIDS ; 16(14): 1849-57, 2002 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-12351944

RESUMO

OBJECTIVES: HIV-specific CD8 T cells play a central role in the immune control of virus replication. To further understand the role of CD8 T cells in clinical settings, there is a need for a diagnostic assay that quantifies HIV-specific CD8 T cells in all HIV-infected individuals. DESIGN: and methods: The CD8VIR (CD8 T cell-mediated virus-specific immune response) assay was designed to mimic viral load rebound by adding replication defective HIV particles to peripheral blood mononuclear cells. Antigen presenting cells processed the virus and presented most of the viral epitopes to T cells. Activated HIV-specific CD8 T cells were quantified by flow cytometry analysis as CD3CD8 IFNgamma producing T cells. RESULTS: The CD8VIR assay reproducibly detected a large proportion of functional HIV-specific CD8 T cells responding to viral load rebound. The whole HIV particle stimulation used in the CD8VIR assay was comparable to the sum of Gag, Pol, Env and Nef stimulations. The percentage of HIV-specific CD8 T cells also significantly correlated with the percentage of Gag-specific cytotoxicity measured by the traditional Cr release assay. HIV-specific CD8 T cells correlated with immune control of HIV in chronically infected patients. CONCLUSIONS: The CD8VIR assay quantifies the majority of HIV-specific CD8 T cells capable of killing HIV-infected cells during viral load rebound. This simple, versatile and reproducible assay can be performed from the specimen submitted for CD4 analysis. Upon clinical validation, the CD8VIR assay can be a new diagnostic tool to predict the control of viral load rebound after treatment interruption.


Assuntos
Antígenos CD8/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por HIV/diagnóstico , Infecções por HIV/imunologia , HIV/imunologia , Bioensaio , Linfócitos T CD8-Positivos/microbiologia , Citometria de Fluxo , Produtos do Gene gag/imunologia , Produtos do Gene nef , Produtos do Gene pol/imunologia , Infecções por HIV/virologia , Humanos , Técnicas In Vitro , Carga Viral , Inativação de Vírus , Produtos do Gene nef do Vírus da Imunodeficiência Humana
17.
AIDS ; 17(1): 43-51, 2003 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-12478068

RESUMO

OBJECTIVE: To study the effect of highly active antiretroviral therapy (HAART) with and without hydroxyurea (HU) on changes in plasma viral load (VL) set-point, and on HIV-1-specific responses, after five cycles of structured treatment interruptions (STI). METHODS: A group of 20 patients taking HAART for chronic HIV infection with VL < 20 copies/ml were randomized to continue HAART or HAART plus HU for 24 weeks followed by five STI cycles. HU was also stopped in cycles 1-3 but continued in cycles 4 and 5. The number of individuals maintaining a VL set-point < 5000 copies/ml during the fifth interruption were determined. RESULTS: VL remained < 5000 copies/ml in eight out of nine patients in the HU group and in four out of ten patients in the HAART group after a median 48 weeks of follow-up after the fifth interruption ( P=0.039). By STI cycle 5, there was a significant increase in the neutralizing activity (NA), in both magnitude and breadth of the total cytotoxic T lymphocyte (CTL) response and in lymphoproliferative response (LPR) from baseline. No significant differences were observed between HAART and HU groups in NA, CTL and LPR at any time-point. There were no differences in the NA titers at any time-point between responder and non-responder patients. There was a trend for higher CTL and LPR levels in responder patients (P= 0.10). CONCLUSIONS: In this randomized, controlled study of STI with cycles of HAART or HAART plus HU, a lower peak VL rebound and a lower VL set-point was achieved in patients continuing HU while other drugs were discontinued. HU did not blunt anti-HIV-1-specific responses; however, control of VL did not correlate with anti-HIV-1-specific cellular immune responses.


Assuntos
Terapia Antirretroviral de Alta Atividade , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Hidroxiureia/uso terapêutico , Replicação Viral/efeitos dos fármacos , Adulto , Fármacos Anti-HIV/efeitos adversos , Fármacos Anti-HIV/uso terapêutico , Terapia Antirretroviral de Alta Atividade/efeitos adversos , Contagem de Linfócito CD4 , Esquema de Medicação , Quimioterapia Combinada , Feminino , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/fisiologia , Humanos , Masculino , Inibidores da Síntese de Ácido Nucleico/uso terapêutico , Carga Viral
18.
Microbes Infect ; 4(2): 207-14, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11880054

RESUMO

Structured treatment interruption (STI) has been investigated for three distinct clinical scenarios: during acute infection with the goal of immune reconstitution and auto immunization; during chronic infection, to decrease the amount and toxicity of antiretroviral drugs; and during virologic failure to restore response to subsequent antiretroviral therapy. The potential costs and benefits of STI should be determined.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/uso terapêutico , Terapia Antirretroviral de Alta Atividade/métodos , Infecções por HIV/tratamento farmacológico , Terapia Antirretroviral de Alta Atividade/economia , Contagem de Linfócito CD4 , Ensaios Clínicos como Assunto , Esquema de Medicação , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Cooperação do Paciente , Risco , Carga Viral
19.
Antivir Ther ; 9(1): 123-32, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15040544

RESUMO

BACKGROUND: Toxicity and other drug adherence-related factors have contributed to decreased compliance to antiretroviral regimens amongst HIV-infected patients. Irregular therapy disruption causes loss of CD4 T cells, onset of drug resistance and rapid rebound of plasma viral load (VL). However, an appropriate choice of drugs and properly scheduled structured treatment interruptions (STIs) may limit VL rebound, maintain CD4 counts and minimize resistance. METHODS: We conducted a clinical study of STIs, RIGHT 901, involving 60 drug-naive patients with chronic HIV infection (CD4 >300, VL >10,000) randomized to receive didanosine-stavudine-indinavir (IDV group) or didanosine-stavudine-hydroxyurea (HU group), for 12 weeks. Subsequently, all patients were randomized again to start STI (short induction) or to continue the therapy for an additional 24 weeks before starting STI (long induction). Both groups underwent four STI cycles and then stopped therapy as long as viraemia remained below 10,000 copies/ml before reinitiating another four cycles of STI. RESULTS: During continuous therapy VLs were suppressed at similar rates in both the HU and IDV groups, while a blunted CD4 count was documented in the HU group. Following the first stop median VL rebounded close to baseline values in both groups, however, during the following STI median VL rebound decreased in the HU group, while in the IDV group VL continued to rebound to values close to baseline, and the difference between the two groups was statistically significant. Moreover, patients treated with HU had a constant and stable CD4 increase during STI, whereas CD4 counts fluctuated in the IDV group, with sharp falls during treatment interruptions and partial CD4 recovery following treatment restart. Even in the presence of IDV resistance predisposing mutations at baseline, no genotypic change in the protease sequence was observed during STI. A relevant mutation in the reverse transcriptase sequence (K70R) emerged in one patient interrupting treatment after 36 weeks of continuous therapy and in one patient after four STI cycles. Side effects (no major events) were similar among groups. CONCLUSIONS: An appropriate choice of STI schedule and regimens containing drugs less prone to resistance and/or able to prevent CD4 fluctuation may contribute to optimizing STI for chronically infected patients with respect to limiting viral rebound, improving CD4 counts and maintaining a resistance profile comparable to continuous highly active antiretroviral therapy.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Farmacorresistência Viral/fisiologia , HIV-1/isolamento & purificação , Carga Viral , Fármacos Anti-HIV/administração & dosagem , Didanosina/uso terapêutico , Esquema de Medicação , Quimioterapia Combinada , Humanos , Hidroxiureia/uso terapêutico , Indinavir/uso terapêutico , Itália , RNA Viral/sangue , RNA Viral/isolamento & purificação , Estavudina/uso terapêutico , Fatores de Tempo , Viremia/epidemiologia
20.
J Immunol Methods ; 276(1-2): 45-57, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12738358

RESUMO

Human immunodeficiency virus (HIV)-specific T cells play a critical role in anti-virus immunity. Therefore, during the clinical development of immune-based therapies, it is important to perform a diagnostic test that rapidly quantifies and characterizes cellular immune responses. For detection of functional HIV-specific CD8(+) cytotoxic T cells (CTL), we used a rapid vaccinia assay that employs recombinant vaccinia virus-infected peripheral blood mononuclear cells (PBMC). This assay was compared with the traditional 51Cr-release CTL assay and with the peptide pool-based enzyme-linked immunospot assay (ELISPOT). We demonstrated a close correlation between these assays by using identical antigens in parallel assays. However, the vaccinia assay was the least expensive and time- and labor-consuming test. Regarding sensitivity, the vaccinia assay was similar to both the peptide pool-based ELISPOT and CTL assays. We showed that human histocompatibility leukocyte antigen (HLA)-Dr(+) cells are responsible for presenting recombinant vaccinia antigens to T cells. The recombinant vaccinia-based assay is a rapid, sensitive and quantitative test and is thus suitable for the detection of functional antigen-specific CD8(+) CTL in large-scale clinical studies.


Assuntos
HIV-1/imunologia , Linfócitos T Citotóxicos/imunologia , Vacínia/genética , Células Apresentadoras de Antígenos/imunologia , Linfócitos B/virologia , Linhagem Celular , Técnicas de Cocultura , Testes Imunológicos de Citotoxicidade , DNA Recombinante/genética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Antígenos HIV/imunologia , Interferon gama/biossíntese , Leucócitos Mononucleares/virologia , Peptídeos/imunologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA