Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Dis Colon Rectum ; 66(3): 360-365, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36599112

RESUMO

BACKGROUND: Anal cancer is associated with high-risk human papillomavirus infection and oncoprotein expression. We have identified several protease inhibitors, used to treat HIV, that decrease oncogene expression. OBJECTIVE: The aim of this project is to determine whether saquinavir, a protease inhibitor, results in a treatment response in anal cancer spheroids. DESIGN: K14E6/E7 transgenic mice (n = 5), which express human papillomavirus 16 oncoproteins E6 and E7 in their epithelium, were treated topically at the anus with a carcinogen, 7,12-dimethylbenz[a]anthracene, to promote anal tumor growth. Tumors were excised and digested, and cells were plated. The tumor cells form 3D multicellular aggregates known as spheroids. SETTINGS: This study was performed in an American Association for Accreditation of Laboratory Animal Care-approved facility. INTERVENTIONS: Spheroids were placed in treatment groups: no treatment, vehicle (dimethyl sulfoxide), and 15 µM saquinavir. Spheroids were imaged immediately pretreatment and 24 hours posttreatment. MAIN OUTCOME MEASURES: Spheroid diameters were measured using ImageJ and mean percent reduction was calculated for each spheroid to determine treatment effect on spheroid growth. Analysis of variance using pairwise comparisons was performed with Fisher protected least significant difference tests. RESULTS: The no-treatment (n = 119 spheroids) and vehicle (n = 126 spheroids) groups demonstrated an increase in spheroid diameter during the treatment period. In contrast, spheroids treated with saquinavir (n = 151 spheroids) demonstrated a statistically significant percent reduction compared to the no-treatment ( p < 0.0001) and vehicle ( p = 0.002) groups. LIMITATIONS: A limitation of these data is that some human error is likely present given that images were analyzed by 3 different scientists. CONCLUSIONS: Saquinavir leads to a statistically significant percent reduction in mice anal tumor spheroid growth ex vivo compared to control groups. Protease inhibitor therapy may be an effective treatment or adjuvant therapy to the Nigro protocol to promote anal cancer tumor regression. See Video Abstract at http://links.lww.com/DCR/C82 . EL USO DEL INHIBIDOR DE LA PROTEASA, SAQUINAVIR, PARA TRATAR LOS ESFEROIDES DEL CNCER ANAL DERIVADOS DE RATONES TRANSGNICOS PARA EL VPH: ANTECEDENTES:El cáncer anal está asociado con la infección por el virus del papiloma humano de alto riesgo y la expresión de oncoproteínas. Hemos identificado varios inhibidores de la proteasa, utilizados para tratar el VIH, que disminuyen la expresión del oncogén.OBJETIVO:El objetivo de este proyecto es determinar si los esferoides de cáncer anal responden al tratamiento con inhibidor de la proteasa, Saquinavir.DISEÑO:Ratones transgénicos K14E6/E7 (n = 5), que expresan las oncoproteínas E6 y E7 del VPH16 en su epitelio, fueron tratados tópicamente en el ano con carcinógeno, 7,12 dimetilbenz[a]antraceno, para promover el crecimiento del tumor anal. Los tumores se extirparon y digirieron, y las células se sembraron en placas. Las células tumorales forman agregados multicelulares tridimensionales, conocidos como esferoides.ESCENARIO:Este estudio se realizó en un centro aprobado por la Asociación Estadounidense para la Acreditación de Cuidado de Animales de Laboratorio.INTERVENCIONES:Se colocaron esferoides en grupos de tratamiento: sin tratamiento, vehículo (sulfóxido de dimetilo) y saquinavir 15 µM. Se tomaron imágenes de los esferoides inmediatamente antes del tratamiento y 24 horas después del tratamiento.PRINCIPALES MEDIDAS DE RESULTADO:Los diámetros de los esferoides se midieron con ImageJ y se calculó el porcentaje medio de reducción de cada esferoide para determinar el efecto del tratamiento sobre el crecimiento de los esferoides. El análisis de varianza mediante comparaciones por pares se realizó con las pruebas de diferencia mínima significativa protegida de Fisher.RESULTADOS:Los grupos sin tratamiento (n =119 esferoides) y vehículo (n=126 esferoides) demostraron un aumento en el diámetro del esferoide durante el período de tratamiento. Por el contrario, los esferoides tratados con saquinavir (n =151 esferoides) demostraron una reducción porcentual estadísticamente significativa en comparación con los grupos sin tratamiento ( p < 0,0001) y con vehículo (p = 0,002).LIMITACIONES:una limitación de estos datos es que es probable que haya algún error humano dado que las imágenes fueron analizadas por tres científicos diferentes.CONCLUSIONES:Saquinavir conduce a una reducción porcentual estadísticamente significativa en el crecimiento de esferoides de tumores anales en ratones ex-vivo en comparación con los grupos de control. La terapia con inhibidores de la proteasa puede ser un tratamiento eficaz o una terapia adyuvante del protocolo Nigro para promover la regresión del tumor del cáncer anal. Consulte Video Resumen en http://links.lww.com/DCR/C82 . (Traducción-Dr. Felipe Bellolio ).


Assuntos
Anti-Infecciosos , Neoplasias do Ânus , Humanos , Camundongos , Animais , Saquinavir/farmacologia , Saquinavir/uso terapêutico , Papillomavirus Humano , Inibidores de Proteases , Camundongos Transgênicos , Neoplasias do Ânus/tratamento farmacológico , Neoplasias do Ânus/patologia , Estudos Retrospectivos
2.
Virology ; 576: 96-104, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36206607

RESUMO

Select protease inhibitors (PI) have been found to be effective in decreasing human papillomavirus oncoprotein expression. This study evaluated whether the topical PI, Saquinavir (SQV), promotes viral clearance in an infectious mouse model with Mus musculus papillomavirus 1 (MmuPV1). NOD scid gamma (NSG) mice were anally infected with ∼4 × 108 viral genome equivalents of MmuPV1 and 120 days post-infection (when majority have high-grade anal dysplasia), began topical treatments: control (mock), 7,12-dimethylbenz(a)anthracene (DMBA) only, once weekly to promote carcinogenesis, 1% SQV only, daily (Monday - Friday), and SQV + DMBA. Viral MmuPV1 load was analyzed from anal lavages pre and post-treatment. Anal tissue was harvested, processed, and evaluated for drug absorption, grade of anal disease, and anal viral RNA. Results suggest that topical SQV promotes decreased viral shedding in female mice treated with SQV.


Assuntos
Infecções por HIV , Inibidores da Protease de HIV , Viroses , Feminino , Camundongos , Humanos , Animais , Saquinavir/farmacologia , Saquinavir/uso terapêutico , Inibidores da Protease de HIV/uso terapêutico , RNA Viral , Carga Viral , Papillomaviridae/genética , Inibidores Enzimáticos , Antracenos
3.
Int J Mol Sci ; 23(20)2022 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-36293096

RESUMO

Prostate and lung cancers are among the most common cancer types, and they still need more therapeutics. For this purpose, saquinavir (SAQ) was tested alone and in combination with 5-fluorouracil (5-FU). PC-3 and A549 cells were exposed to increasing concentrations of both drugs alone or in combination, with simultaneous or sequential administration. Cell viability was obtained using the MTT assay and synergism values using CompuSyn software. Results showed that SAQ was the more cytotoxic of both drugs in PC-3 cells, while 5-FU was the most cytotoxic in A549 cells. When these drugs were used in combination, the more synergistic combination in PC-3 cells was the IC50 of SAQ with various concentrations of 5-FU, particularly when 5-FU was only applied 24 h later. Meanwhile for A549 the most promising combination was 5-FU with delayed SAQ, but with a weaker effect than all combinations demonstrated in PC-3 cells. These results demonstrate that SAQ could be used as a new repurposed drug for the treatment of prostate cancer and this treatment potential could be even greater if SAQ is combined with the anticancer drug 5-FU, while for lung cancer it is not as efficient and, therefore, not of as much interest.


Assuntos
Antineoplásicos , Neoplasias Pulmonares , Masculino , Humanos , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Saquinavir/farmacologia , Saquinavir/uso terapêutico , Próstata , Antivirais/uso terapêutico , Linhagem Celular Tumoral , Reposicionamento de Medicamentos , Sinergismo Farmacológico , Antineoplásicos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico
4.
Plast Reconstr Surg ; 150(6): 1264e-1274e, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36112847

RESUMO

BACKGROUND: Using immunomodulatory methods to address the challenging issue of craniofacial bone repair may be a potentially effective approach. The protease inhibitor saquinavir has been shown to inhibit the inflammatory response by targeting the toll-like receptor 4/myeloid differentiation primary response complex. Independently, inhibition of toll-like receptor 4 or myeloid differentiation primary response led to enhanced skull bone repair. Therefore, the authors aimed to investigate the effects of saquinavir on skull bone healing. METHODS: The effects of saquinavir on skull bone healing were assessed by means of gene expression, histology, immunohistochemistry, and tomography in a mouse calvarial defect model. Subsequently, the role of saquinavir in cell viability, migration, and osteogenic and osteoclastogenic differentiation was also evaluated in vitro. RESULTS: One-week saquinavir administration improved skull bone healing based on micro-computed tomographic and histomorphometric analyses. Compared to the vehicle control, 1-week saquinavir treatment (1) enhanced osteoclast infiltration (tartrate-resistant acid phosphatase staining) at day 7, but not at days 14 and 28; (2) induced more CD206 + M2 macrophage infiltration, but not F4/80 + M0 macrophages at days 7, 14, and 28; and (3) elevated osteoclastogenic gene RANKL (quantitative polymerase chain reaction) expression and other osteogenic and cytokine expression. Furthermore, in vitro data showed that saquinavir administration did not influence MC3T3-E1 cell migration or mineralization, whereas higher concentrations of saquinavir inhibited cell viability. Saquinavir treatment also enhanced the osteoclastic differentiation of bone marrow-derived precursors, and partially reversed high-mobility group box 1-driven osteoclastogenesis inhibition and elevated proinflammatory cytokine expression. CONCLUSION: The improved skull bone repair following short-term saquinavir treatment may involve enhanced osteoclastogenesis and modulated inflammatory response following skull injury. CLINICAL RELEVANCE STATEMENT: The authors' work demonstrates improved skull bone healing by short-term application of saquinavir, a drug traditionally used in the treatment of acquired immunodeficiency syndrome. As such, saquinavir may be repurposed for skeletal repair.


Assuntos
Inibidores da Protease de HIV , Saquinavir , Camundongos , Animais , Saquinavir/farmacologia , Saquinavir/metabolismo , Saquinavir/uso terapêutico , Inibidores da Protease de HIV/farmacologia , Inibidores da Protease de HIV/metabolismo , Inibidores da Protease de HIV/uso terapêutico , Receptor 4 Toll-Like/fisiologia , Osteogênese , Crânio/lesões
5.
Biomolecules ; 12(7)2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35883499

RESUMO

Saquinavir was the first protease inhibitor developed for HIV therapy, and it changed the standard of treatment for this disease to a combination of drugs that ultimately led to increased survival of this otherwise deadly condition. Inhibiting the HIV protease impedes the virus from maturing and replicating. With this in mind, since the start of the COVID-19 outbreak, the research for already approved drugs (mainly antivirals) to repurpose for treatment of this disease has increased. Among the drugs tested, saquinavir showed promise in silico and in vitro in the inhibition of the SARS-CoV-2 main protease (3CLpro). Another field for saquinavir repurposing has been in anticancer treatment, in which it has shown effects in vitro and in vivo in several types of cancer, from Kaposi carcinoma to neuroblastoma, demonstrating cytotoxicity, apoptosis, inhibition of cell invasion, and improvement of radiosensibility of cancer cells. Despite the lack of follow-up in clinical trials for cancer use, there has been a renewed interest in this drug recently due to COVID-19, which shows similar pharmacological pathways and has developed superior in silico models that can be translated to oncologic research. This could help further testing and future approval of saquinavir repurposing for cancer treatment.


Assuntos
Tratamento Farmacológico da COVID-19 , Infecções por HIV , Inibidores da Protease de HIV , Neoplasias , Infecções por HIV/tratamento farmacológico , Inibidores da Protease de HIV/farmacologia , Inibidores da Protease de HIV/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , SARS-CoV-2 , Saquinavir/farmacologia , Saquinavir/uso terapêutico
6.
Travel Med Infect Dis ; 35: 101646, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32294562

RESUMO

BACKGROUND: The COVID-19 has now been declared a global pandemic by the World Health Organization. There is an emergent need to search for possible medications. METHOD: Utilization of the available sequence information, homology modeling, and in slico docking a number of available medications might prove to be effective in inhibiting the SARS-CoV-2 two main drug targets, the spike glycoprotein, and the 3CL protease. RESULTS: Several compounds were determined from the in silico docking models that might prove to be effective inhibitors for SARS-CoV-2. Several antiviral medications: Zanamivir, Indinavir, Saquinavir, and Remdesivir show potential as and 3CLPRO main proteinase inhibitors and as a treatment for COVID-19. CONCLUSION: Zanamivir, Indinavir, Saquinavir, and Remdesivir are among the exciting hits on the 3CLPRO main proteinase. It is also exciting to uncover that Flavin Adenine Dinucleotide (FAD) Adeflavin, B2 deficiency medicine, and Coenzyme A, a coenzyme, may also be potentially used for the treatment of SARS-CoV-2 infections. The use of these off-label medications may be beneficial in the treatment of the COVID-19.


Assuntos
Betacoronavirus/química , Infecções por Coronavirus/virologia , Cisteína Endopeptidases/química , Descoberta de Drogas/métodos , Pneumonia Viral/virologia , Glicoproteína da Espícula de Coronavírus/química , Proteínas não Estruturais Virais/química , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/química , Monofosfato de Adenosina/uso terapêutico , Alanina/análogos & derivados , Alanina/química , Alanina/uso terapêutico , Sítios de Ligação , COVID-19 , Proteases 3C de Coronavírus , Infecções por Coronavirus/tratamento farmacológico , Inibidores da Protease de HIV/química , Inibidores da Protease de HIV/uso terapêutico , Humanos , Indinavir/química , Indinavir/uso terapêutico , Simulação de Acoplamento Molecular , Uso Off-Label , Pandemias , Pneumonia Viral/tratamento farmacológico , SARS-CoV-2 , Saquinavir/química , Saquinavir/uso terapêutico , Glicoproteína da Espícula de Coronavírus/antagonistas & inibidores , Homologia Estrutural de Proteína , Proteínas não Estruturais Virais/antagonistas & inibidores , Zanamivir/química , Zanamivir/uso terapêutico , Tratamento Farmacológico da COVID-19
7.
Mol Med ; 23: 92-100, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28332696

RESUMO

The inflammatory pathways that drive the development of intimal hyperplasia (IH) following arterial injury are not fully understood. We hypothesized that the lysosomal cysteine protease cathepsin L activates processes leading to IH after arterial injury. Using a mouse model of wire-induced carotid artery injury we showed that cathepsin L activity peaks at day 7 and remains elevated to 28 days. The genetic deletion of cathepsin L prevented IH and monocyte recruitment in the carotid wall. The injury-induced increases in cathepsin L mRNA and activity were mitigated in mice with myeloid-specific deletion of toll like receptor 4 (TLR4) or myeloid differentiation primary response gene 88 (MyD88). We further discovered that a HIV-protease inhibitor saquinavir (SQV), which is known to block recombinant mouse cathepsin L activity in vitro, prevented IH after arterial injury. SQV also suppressed LPS (TLR4 agonist) induced monocyte adhesion to endothelial monolayers. These findings establish cathepsin L as a critical regulator of the inflammation that leads to IH and that the TLR4- MyD88 pathway in myeloid lineages regulates cathepsin L expression in the vessel wall following wire injury. The FDA approved drug, SQV blocks IH though mechanisms that may include the suppression of cathepsin L.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Catepsina L/metabolismo , Hiperplasia/metabolismo , Túnica Íntima/patologia , Animais , Lesões das Artérias Carótidas/tratamento farmacológico , Catepsina L/genética , Células Cultivadas , Inibidores da Protease de HIV/uso terapêutico , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Hiperplasia/tratamento farmacológico , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Saquinavir/uso terapêutico , Receptor 4 Toll-Like/metabolismo
8.
Mediators Inflamm ; 2017: 7083528, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29440779

RESUMO

Liver ischemia and reperfusion (I/R) induce local and distant tissue injuries, contributing to morbidity and mortality in a wider range of pathologies. This is especially seen under uncontrolled aseptic inflammatory conditions, leading to injury of remote organs, such as lung injury, and even failure. Saquinavir (SQV) is a kind of HIV protease inhibitor that possesses an anti-inflammatory property. In this study, we investigated whether SQV suppresses Toll-like receptor 4- (TLR4-) dependent signaling pathways of high-mobility group box 1 (HMGB1) and P38/JNK, conferring protection against murine liver I/R-induced lung injury. To investigate our hypothesis, C57BL/6 mice and TLR4 knockout mice (TLR4-/-) were used to perform the study. SQV administration markedly attenuated remote lung tissue injury after 1-hour ischemia and 6-hour reperfusion of the liver. To our expectation, SQV attenuated I/R-induced lung edema, hyperpermeability, and pathological injury. The beneficial effects of SQV were associated with decreased levels of circulating and lung tissue inflammatory cytokines, such as IL-6, IL-1ß, TNF-α, and iNOS. The protective effect of SQV was also associated with decreased lung tissue expression of HMGB1, TLR-4, and p-P38/JNK, but not p-ERK in wild-type liver I/R mice. Overall, this study demonstrated a new role of SQV, facilitating negative regulation of HMGB1- and P38/JNK-mediated TLR-4-dependent signaling pathways, conferring protection against liver I/R-induced lung injury.


Assuntos
Proteína HMGB1/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Fígado/irrigação sanguínea , Lesão Pulmonar/tratamento farmacológico , Traumatismo por Reperfusão/tratamento farmacológico , Saquinavir/uso terapêutico , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Citocinas/fisiologia , Lesão Pulmonar/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão/imunologia , Isquemia Quente
9.
Int J Cancer ; 140(8): 1713-1726, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-27870005

RESUMO

The possible use of HIV protease inhibitors (HIV-PI) as new therapeutic option for the treatment of cancer primarily originated from their success in treating HIV-related Kaposi's sarcoma (KS). While these findings were initially attributed to immune reconstitution and better control of oncogenic viral infections, the number of reports on solid tumors, KS, lymphoma, fibrosarcoma, multiple myeloma and prostate cancer suggest other mechanisms for the anti-neoplastic activity of PIs. However, a major drawback for the possible adoption of HIV-PIs in the therapy of cancer relies on their relatively weak anticancer potency and important side effects. This has propelled several groups to generate derivatives of HIV-PIs for anticancer use, through modifications such as attachment of different moieties, ligands and transporters, including saquinavir-loaded folic acid conjugated nanoparticles and nitric oxide (NO) derivatives of HIV-PIs. In this article, we discuss the current preclinical and clinical evidences for the potential use of HIV-PIs, and of novel derivatives, such as saquinavir-NO in the treatment of cancer.


Assuntos
Infecções por HIV/tratamento farmacológico , Inibidores da Protease de HIV/uso terapêutico , Neoplasias/tratamento farmacológico , Sarcoma de Kaposi/tratamento farmacológico , Terapia Antirretroviral de Alta Atividade , Infecções por HIV/complicações , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/patogenicidade , Humanos , Neoplasias/complicações , Neoplasias/virologia , Óxido Nítrico/metabolismo , Saquinavir/análogos & derivados , Saquinavir/uso terapêutico , Sarcoma de Kaposi/complicações , Sarcoma de Kaposi/virologia
10.
J Exp Clin Cancer Res ; 32: 38, 2013 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-23759068

RESUMO

BACKGROUND: Saquinavir, a protease inhibitor utilized in HIV infection, shows antitumor activity in various experimental models. In previous studies performed in our laboratory the drug was found to induce a substantial increase of telomerase activity in normal peripheral blood mononuclear cells. Aim of the present investigation was to test whether saquinavir was able to increase telomerase activity and the expression of the catalytic subunit of telomerase, hTERT, in human malignant hematopoietic cells. METHODS: Human Jurkat CD4+ T cell leukaemia cell line was used throughout the present study. The antiproliferative effect of saquinavir was tested by the MTT assay. Telomerase activity was determined according to the telomeric repeat amplification protocol. The expression of hTERT mRNA was semi-quantitative evaluated by RT-PCR amplification and quantitative Real Time PCR. The binding of the transcription factor c-Myc to its specific E-Box DNA binding-site of hTERT promoter was analyzed by Electophoretic Mobility Shift Assay (EMSA). The amount of c-Myc in cytoplasm and nucleus of leukemia cells was determined by Western Blot analysis, and c-Myc down-regulation was obtained by siRNA transfection. RESULTS: Saquinavir produced a substantial increase of telomerase activity in Jurkat cells in vitro without increasing but rather reducing target cell proliferation rate. Telomerase up-regulation appeared to be the result of enhanced expression of hTERT. Saquinavir-mediated up-regulation of hTERT gene was the result of the increased binding of proteins to the E-Box sequence of the promoter. Moreover, saquinavir amplified the expression of c-Myc especially in the nuclear cell fraction. The direct influence of saquinavir on this transcription factor was also demonstrated by the antagonistic effect of the drug on siRNA induced c-Myc suppression. Since c-Myc is the main responsible for hTERT transcription, these findings suggest that the main mechanism underlying saquinavir-induced telomerase activation is mediated by c-Myc up-regulation. CONCLUSIONS: Saquinavir augments hTERT expression while inhibiting leukemic cell growth. Experimental evidences show that this effect is mediated by saquinavir-influenced increase of c-Myc levels. This could have relevance in terms of enhanced hTERT-dependent tumor cell immunogenicity and suggests new paharmacological approaches interfering with c-Myc dependent pathways.


Assuntos
Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Inibidores da Protease de HIV/farmacologia , Leucemia de Células T/genética , Leucemia de Células T/metabolismo , Saquinavir/farmacologia , Telomerase/genética , Telomerase/metabolismo , Domínio Catalítico/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inibidores da Protease de HIV/uso terapêutico , Humanos , Células Jurkat , Leucemia de Células T/tratamento farmacológico , Proteínas Proto-Oncogênicas c-myc/metabolismo , Saquinavir/uso terapêutico , Telomerase/química , Transcrição Gênica/efeitos dos fármacos
11.
Ter Arkh ; 85(11): 16-20, 2013.
Artigo em Russo | MEDLINE | ID: mdl-24432594

RESUMO

AIM: To retrospectively analyze the efficacy and safety of ritonavir-boosted saquinavir (SQV) in the perinatal prevention of HIV infection in pregnant women in the Ulyanovsk Region. MATERIALS AND METHODS: The outpatient cards of HIV-positive pregnant women who received ritonavir-boosted SQV in a dose of 1000/100 mg bid in combination with zidovudine or stavudine and lamivudine for the perinatal prevention of HIV infection in 2010-2011 were retrospectively analyzed. The main efficiency criterion was a proportion of children taken off a regular medical check-up in terms of perinatal contact as healthy at 18 months post-birth. The additional criteria (end points) were changes in viral load and CD4 cell count at 4 and 8-12 weeks after initiation of prevention. RESULTS: The analysis included data on 98 HIV-positive pregnant women. The history of HIV infection was 3 years or less in 63.2% of the patients. Stage 3 and 4A HIV infection was stated in 87 and 13% of the women, respectively. Treatment with ritonavir-boosted SQV + 2 nucleotide reverse transcriptase inhibitors was started in 84.8% of the women at 15-28 weeks' gestation and in 12.2% of cases in earlier periods. Four weeks after initiation of highly active antiretroviral therapy, the viral load showed a 1-4 log10 decrease. By that time and at 8-12 weeks, it was undetectable (< 500 copies/ml) in 75 (76.5%) and 82 (83.6%) women, respectively. With a decrease in the viral load, CD4 cell count increased by 100-500 per mm3 (mean 160 per mm3). The efficiency of prevention of perinatal HIV transmission was 100%. No serious adverse events were observed. CONCLUSION: The use of ritonavir-boosted SQV in combination with two nucleotide reverse transcriptase inhibitors is an effective and safe chemoprevention regimen for perinatal HIV transmission in pregnant women.


Assuntos
Infecções por HIV/tratamento farmacológico , HIV , Assistência Perinatal/métodos , Complicações Infecciosas na Gravidez , Saquinavir/uso terapêutico , Terapia Antirretroviral de Alta Atividade/métodos , Feminino , Seguimentos , Infecções por HIV/epidemiologia , Infecções por HIV/transmissão , Inibidores da Protease de HIV/uso terapêutico , Humanos , Incidência , Gravidez , Estudos Retrospectivos , Federação Russa/epidemiologia , Resultado do Tratamento
12.
Mol Pharmacol ; 82(4): 700-10, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22798453

RESUMO

We have examined the influence of the nitric oxide (NO)-modified anti-inflammatory drug (S,R)-3-phenyl-4,5-dihydro-5-isoxasole acetic acid (VGX-1027) named GIT-27NO or the NO-modified antiviral drug saquinavir (Saq) named Saq-NO on two colon cancer cell lines, mouse CT26CL25 and human HCT116. The effects of the drugs on cell viability, apoptosis, proliferation, and metastatic potential were analyzed. The release of NO and oxygen and nitrogen species was also determined. The efficacy of the drugs was evaluated in vivo in BALB/c mice injected with CT26CL25 cells. Both agents suppressed the growth of colon cancer cells in vitro and reduced tumor volume in syngeneic BALB/c mice. However, their mechanisms of action were different because GIT-27NO released larger amounts of nitrite than Saq-NO in cell cultures and its antitumor action depended on the intracellular NO release inside the cells. On the contrary, Saq-NO released barely detectable amounts of NO and its antitumor action was NO-independent. In fact, cotreatment with an NO-peroxynitrite scavenger revealed that GIT-27NO but not Saq-NO acts through peroxynitrite-mediated cell destruction. At the cellular level, GIT-27NO prevalently induced proapoptotic signals followed by caspase-dependent apoptosis. In contrast, Saq-NO blocked cell proliferation, changed the adhesive, migratory, and invasive properties of the cells, and decreased metastatic potential in vivo. In conclusion, differences in NO release and oxidative stress generation between GIT-27NO and Saq-NO resulted in different mechanisms that caused cell death.


Assuntos
Acetatos/farmacologia , Antineoplásicos/farmacologia , Neoplasias do Colo/tratamento farmacológico , Óxido Nítrico/química , Oxazóis/farmacologia , Saquinavir/farmacologia , Acetatos/química , Acetatos/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Transplante de Neoplasias , Óxido Nítrico/fisiologia , Oxazóis/química , Oxazóis/uso terapêutico , Saquinavir/química , Saquinavir/uso terapêutico
13.
Antivir Ther ; 16(8): 1237-47, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22155905

RESUMO

BACKGROUND: Matrix metalloproteinases (MMPs) and their endogenous tissue inhibitors (TIMPs) are central to tissue remodelling during HIV-HCV infection. Here, we assess the potential for antiviral therapy to modulate MMP abundance in THP-1 monocyte/macrophages and LX-2 hepatic stellate cells, and in a coinfected patient cohort. METHODS: THP-1 and LX-2 cells were treated with ribavirin (RBV)/interferon-α (IFN-α) and select HIV antivirals. Venous blood was reserved from HIV-HCV-coinfected patients, HIV- and HCV-monoinfected patients, and healthy controls, with the HIV-HCV cohort being sampled again at day 3 and 14 subsequent to the start of combination therapy with RBV/pegylated IFN-α. Samples were subjected to gelatin zymography, real-time RT-PCR and/or ELISA, where appropriate. RESULTS: RBV/IFN-α decreased MMP-9 activity, and increased MMP-9 mRNA and protein expression in THP-1 cells, but not in LX-2 cells. Decreases in MMP-9 activity were mediated by IFN-α, which also attenuated RBV induction of MMP-9 activity and protein expression in THP-1 cells. Saquinavir and lopinavir, HIV protease inhibitors, reduced MMP-9 activity in THP-1 and LX-2 cells, respectively. Plasma MMP-9 activity and expression was higher in HIV-HCV and HIV patients compared with HCV patients and healthy controls. MMP-2 and TIMP-2 levels were similar in all groups. RBV/pegylated IFN-α decreased plasma MMP-9 abundance in HIV-HCV patients. CONCLUSIONS: These data demonstrate that RBV/pegylated IFN-α reduce plasma MMP-9 abundance in vivo and may reduce its activity in vitro through immune cells, such as monocyte/macrophages, rather than hepatic stellate cells. The results of this study indicate that such therapy may mediate tissue remodelling associated with HIV-HCV coinfection through effects on MMP-9.


Assuntos
Antivirais/administração & dosagem , Infecções por HIV/tratamento farmacológico , HIV/efeitos dos fármacos , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/tratamento farmacológico , Adulto , Antivirais/uso terapêutico , Linhagem Celular , Estudos de Coortes , Coinfecção , Feminino , HIV/fisiologia , Infecções por HIV/sangue , Infecções por HIV/complicações , Infecções por HIV/virologia , Hepacivirus/fisiologia , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Hepatite C Crônica/sangue , Hepatite C Crônica/complicações , Hepatite C Crônica/virologia , Humanos , Interferon alfa-2 , Interferon-alfa/administração & dosagem , Interferon-alfa/uso terapêutico , Irlanda , Lopinavir/administração & dosagem , Lopinavir/uso terapêutico , Masculino , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , RNA Viral , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Ribavirina/administração & dosagem , Ribavirina/uso terapêutico , Saquinavir/administração & dosagem , Saquinavir/uso terapêutico , Inibidor Tecidual de Metaloproteinase-2/sangue , Reino Unido , Carga Viral/efeitos dos fármacos
15.
Cell Cycle ; 10(3): 492-9, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21270522

RESUMO

The NO-derivative of the HIV protease inhibitor saquinavir (Saq-NO) is a nontoxic variant of the parental drug with enhanced anticancer activity on several cell lines. However, it is still unclear whether the p53 status of the target cell might influence the sensitivity to Saq-NO. In this study we evaluated the in vitro and in vivo activity of Saq-NO on the p53-deficient hormone resistant prostate cancer PC-3 cells. We demonstrate that the absence of functional p53 is not essential for the capacity of Saq-NO to reduce prostate cancer cell growth. In contrast to its previously described cytostatic action in B16 and C6 cell lines, Saq-NO exerted cytotoxic effects in PC-3 cells leading to dominant induction of apoptosis and enhanced production of proapoptotic Bim. In addition, differently from saquinavir, Saq-NO restored TRAIL sensitivity that was correlated with increased expression of DR5 independent from ROS/RNS production and YY1 repression. NF-κB activation may be responsible of the Saq-NO induced DR5 expression. Moreover, Saq-NO but not saquinavir, exerted synergistic activity with conventional cytostatic therapy. In agreement with these in vitro studies, Saq-NO inhibited the in vivo growth of PC-3 cells xenotransplants to a greater extent than the parental compound. Taken together, these data indicate that Saq-NO possesses powerful and suitable in vitro and in vivo chemotherapeutic potential to be further studied as a novel drug for the treatment of prostate cancer in the clinical setting.


Assuntos
Antineoplásicos/farmacologia , Inibidores de Proteases/farmacologia , Saquinavir/análogos & derivados , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Humanos , Masculino , Camundongos , Camundongos Nus , Paclitaxel/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Inibidores de Proteases/uso terapêutico , Saquinavir/farmacologia , Saquinavir/uso terapêutico , Transplante Heterólogo , Proteína Supressora de Tumor p53/genética
16.
Biol Pharm Bull ; 32(12): 2002-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19952419

RESUMO

The ATP binding cassette (ABC)-transporters are energy dependent efflux pumps which regulate the pharmacokinetics of both anti-cancer chemotherapeutic agents, e.g. taxol, and of human immunodeficiency virus-1 (HIV-1) protease inhibitors (HPIs), e.g. saquinavir. Increased expression of several ABC-transporters, especially P-glycoprotein (P-gp) and multidrug resistance protein 2 (MRP2), are observed in multidrug resistant (MDR) tumor cells and on HIV-1 infected lymphocytes. In addition, due to their apical expression on vascular endothelial barriers, both P-gp and MRP2 are of crucial importance towards dictating drug access into sequestered tissues. However, although a number of P-gp inhibitors are currently in clinical trials, possible inhibitors of MRP2 are not being thoroughly investigated. The experimental leukotriene receptor antagonist (LTRA), MK-571 is known to be a potent inhibitor of MRP transporters. Using the MRP2 over-expressing Madin-Darby canine kidney cell line, MDCKII-MRP2, we evaluated whether the clinically approved LTRAs, e.g. montelukast (Singulair) and zafirlukast (Accolate), can similarly suppress MRP2-mediated efflux. We compared the efficacy of increasing concentrations (20-100 microM) of MK-571, montelukast, and zafirlukast, in suppressing the efflux of calcein-AM, a fluorescent MRP substrate, and the radiolabeled [(3)H-] drugs, taxol and saquinavir. Montelukast was the most potent inhibitor (p<0.01) of MRP2-mediated efflux of all three substrates. Montelukast also increased (p<0.01) the duration of intracellular retention of both taxol and saquinavir. More than 50% of the drugs were retained in cells even after 90 min post removal of montelukast from the medium. Our findings implicate that montelukast, a relatively safe anti-asthmatic agent, may be used as an adjunct therapy to suppress the efflux of taxol and saquinavir from MRP2 overexpressing cells.


Assuntos
Acetatos/farmacologia , Antineoplásicos Fitogênicos/farmacocinética , Inibidores da Protease de HIV/farmacocinética , Antagonistas de Leucotrienos/farmacologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Paclitaxel/farmacocinética , Quinolinas/farmacologia , Saquinavir/farmacocinética , Animais , Antiasmáticos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Quimioterapia Adjuvante , Ciclopropanos , Cães , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Farmacorresistência Viral/efeitos dos fármacos , Fluoresceínas/metabolismo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo , Inibidores da Protease de HIV/uso terapêutico , Indóis , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Paclitaxel/uso terapêutico , Fenilcarbamatos , Propionatos/farmacologia , Saquinavir/uso terapêutico , Sulfetos , Sulfonamidas , Fatores de Tempo , Compostos de Tosil/farmacologia
17.
Lancet Oncol ; 10(1): 61-71, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19111246

RESUMO

The use of anti-HIV drugs as cancer treatments is not new. Azidothymidine was studied as an antineoplastic in the 1990s, but despite promising in vitro data, clinical trials showed little antitumour activity. HIV protease inhibitors were developed in the early 1990s, and their subsequent incorporation into highly active antiretroviral therapy (HAART) has profoundly changed the natural history of HIV infection. The potential antitumour properties of these drugs have been investigated because of their success in treating HIV-related Kaposi's sarcoma. HAART's effects on Kaposi's sarcoma did not always correlate with immune reconstitution, and activity against other solid and haematological malignancies has been established. Inhibition of tumour-cell invasion and angiogenesis were properties first ascribed to inhibition of HIV protease; however, they have pleiotropic antitumour effects, including inhibition of inflammatory cytokine production, proteasome activity, cell proliferation and survival, and induction of apoptosis. HIV protease inhibitors are thus a new class of anticancer drugs with multiple effects, and other anti-HIV drugs might hold similar promise.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Neoplasias/tratamento farmacológico , Terapia Antirretroviral de Alta Atividade , Cidofovir , Citosina/análogos & derivados , Citosina/uso terapêutico , Inibidores da Protease de HIV/uso terapêutico , Humanos , Indinavir/uso terapêutico , Nelfinavir/uso terapêutico , Organofosfonatos/uso terapêutico , Receptores CXCR4/antagonistas & inibidores , Inibidores da Transcriptase Reversa/uso terapêutico , Ritonavir/uso terapêutico , Saquinavir/uso terapêutico , Zidovudina/uso terapêutico
18.
Curr HIV Res ; 6(6): 555-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18991621

RESUMO

Antiretroviral therapy in human immunodeficiency virus infection is occasionally associated with poor immunologic responses despite full suppression of viral replication. As some combinations of nucleoside analogues (NA) have been associated with paradoxical depletion of CD4(+) T- cells, we postulated that depleting the antiretroviral regimen of NAs would improve quantitative immunological parameters. In a longitudinal prospective study we quantified CD4(+) T-cells after removing NAs from antiretroviral therapy. The NA for regimen consisted of atazanavir (300 mg qd), saquinavir (1000 mg bid), and ritonavir (100mg qd) in 14 patients with immunologic failure despite undetectable plasma HIV-RNA (CD4(+) T-cells < 250 cells/microL (<17%) HIV RNA, <= 50 copies/mL). Additionally, we assessed the state of immunologic activation markers (CD38(+)HLA-DR(+) on CD4(+) and CD8(+) T-cells) by flow cytometry. The regimen was well tolerated. During the 48 week study CD4(+) T-cell counts improved significantly (mean and +/- SEM [standard error of mean], baseline: 174/microL (12.4%) [15, 5.8%], week 24: 232/microL (14%) [26, 5.3%], week 48: 267/microL (15.4%) [34, 4.3%]) with preservation of full viral suppression (p<0.05). Activation parameters of CD4(+) T-cells, but not of CD8(+) T-cells, decreased significantly. This treatment strategy may represent an option for patients with poor immunologic responses to antiretroviral therapy despite undetectable viremia.


Assuntos
Terapia Antirretroviral de Alta Atividade/métodos , Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Inibidores da Protease de HIV/uso terapêutico , Carga Viral , ADP-Ribosil Ciclase 1/análise , Adulto , Sulfato de Atazanavir , Contagem de Linfócito CD4 , Feminino , Citometria de Fluxo , Inibidores da Protease de HIV/administração & dosagem , Inibidores da Protease de HIV/efeitos adversos , Antígenos HLA-DR/análise , Humanos , Estudos Longitudinais , Masculino , Glicoproteínas de Membrana/análise , Pessoa de Meia-Idade , Oligopeptídeos/administração & dosagem , Oligopeptídeos/efeitos adversos , Oligopeptídeos/uso terapêutico , Estudos Prospectivos , Piridinas/administração & dosagem , Piridinas/efeitos adversos , Piridinas/uso terapêutico , Ritonavir/administração & dosagem , Ritonavir/efeitos adversos , Ritonavir/uso terapêutico , Saquinavir/administração & dosagem , Saquinavir/efeitos adversos , Saquinavir/uso terapêutico , Subpopulações de Linfócitos T/imunologia
19.
J Antimicrob Chemother ; 62(6): 1344-55, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18824460

RESUMO

OBJECTIVES: The aim of this study was to develop and validate a population pharmacokinetic model in order to describe ritonavir-boosted saquinavir concentrations dosed twice and once daily in human immunodeficiency virus (HIV)-infected patients from the UK, Uganda and Thailand and to identify factors that may influence saquinavir pharmacokinetics. METHODS: Pharmacokinetic data from 10 clinical studies were combined. Non-linear mixed effects modelling (NONMEM version V) was applied to determine the saquinavir pharmacokinetic parameters, interindividual/interoccasion variability (IIV/IOV) and residual error. Various covariates potentially related to saquinavir pharmacokinetics were explored, and the final model was validated by means of 95% prediction interval and testing the predictive performance of the model with data not included in the model-building process. RESULTS: Ninety-seven patients were included from the UK (n = 52), Uganda (n = 18) and Thailand (n = 27), contributing 347 saquinavir profiles (1-14 profiles per patient). A one-compartment model with zero-order absorption and lag-time best described the data with IIV/IOV on apparent oral clearance (CL/F) and volume of distribution (V/F) and with IIV on duration and absorption lag-time. The ritonavir area under the curve over the dosing interval was significantly associated with saquinavir CL/F and V/F. A typical patient from the UK had approximately 1.5- and 3-fold higher saquinavir CL/F compared with patients from Uganda (89.0 versus 49.8 L/h) and Thailand (89.0 versus 26.7 L/h), respectively. CONCLUSIONS: A model to characterize ritonavir-boosted saquinavir pharmacokinetics in HIV-infected adults has been developed and validated. The model could be used for dosage adaptation following therapeutic drug monitoring and to assess patients' suitability for once-daily boosted saquinavir therapy.


Assuntos
Fármacos Anti-HIV/farmacocinética , Fármacos Anti-HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , Ritonavir/farmacocinética , Ritonavir/uso terapêutico , Saquinavir/farmacocinética , Saquinavir/uso terapêutico , Adulto , Fármacos Anti-HIV/administração & dosagem , Área Sob a Curva , Feminino , HIV/efeitos dos fármacos , Humanos , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Modelos Teóricos , Ritonavir/administração & dosagem , Saquinavir/administração & dosagem , Soro/química , Tailândia , Uganda , Reino Unido
20.
Pediatr Infect Dis J ; 27(7): 623-8, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18520443

RESUMO

OBJECTIVES: To assess the 48-week efficacy, safety, pharmacokinetics, and resistance of double boosted protease inhibitors (PI), saquinavir (SQV), and lopinavir/ritonavir (LPV/r), in children who have failed nucleoside reverse transcription inhibitors /non-nucleoside reverse transcription inhibitors-based regimens. METHODS: Fifty children at 2 sites in Thailand were treated with standard dosing of SQV and LPV/r. CD4, HIV-RNA viral load (VL), plasma drug concentrations and safety laboratory evaluations were monitored. Virologic failure was defined as having 2 consecutive VL >400 copies/mL after week 12 of therapy. Intention to treat analysis was performed. RESULTS: Baseline data were a median age of 9.3 years (interquartile range [IQR]: 7.1-11.2), Center for Disease Control and Prevention (CDC) classification N:A:B:C 4%:14%:68%:14%, VL 4.8 log10 (IQR: 4.5-5.1), CD4 7% (IQR: 3-9.5). At 48 weeks, 3 had died of bacterial infection but no cases had progressed CDC classification. Median CD4% rise was 9 (IQR: 5-16) and median HIV RNA reduction was -2.8 log10 (IQR: -3.2 to -1.4), both P < 0.001. Thirty-nine (78%) and 32 (64%) children had VL <400 and <50 with significant differences between the 2 sites. Five children (10%) had VL failure as a result of poor adherence to the drug regimen but no one had major PI mutations. Median serum cholesterol and triglyceride increased significantly (+35 mg/dL, +37 mg/dL, respectively, both P < 0.001). Mean minimum plasma concentrations (Cmin) of LPV and SQV were 4.6 and 1.24 mg/L, respectively. CONCLUSIONS: Double boosted SQV/LPV/r resulted in significant CD4 rise and VL decline at 48 weeks. Hyperlipidemia was common. Cmin of both PIs exceeded therapeutic concentrations. Poor adherence caused failure in 10%. No major PI mutations were found.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , Inibidores da Protease de HIV/uso terapêutico , Pirimidinonas/uso terapêutico , Ritonavir/uso terapêutico , Saquinavir/uso terapêutico , Fármacos Anti-HIV/efeitos adversos , Fármacos Anti-HIV/farmacocinética , Análise Química do Sangue , Contagem de Linfócito CD4 , Criança , Colesterol/sangue , Farmacorresistência Viral , Feminino , HIV/efeitos dos fármacos , Inibidores da Protease de HIV/efeitos adversos , Inibidores da Protease de HIV/farmacocinética , Humanos , Hiperlipidemias/induzido quimicamente , Lopinavir , Masculino , Estudos Prospectivos , Pirimidinonas/efeitos adversos , Pirimidinonas/farmacocinética , Ritonavir/efeitos adversos , Ritonavir/farmacocinética , Saquinavir/efeitos adversos , Saquinavir/farmacocinética , Tailândia , Recusa do Paciente ao Tratamento , Triglicerídeos/sangue , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA