Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 459
Filter
1.
Bioorg Chem ; 136: 106549, 2023 07.
Article in English | MEDLINE | ID: mdl-37119785

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1), a lentivirus that causes acquired immunodeficiency syndrome (AIDS), poses a serious threat to global public health. Since the advent of the first drug zidovudine, a number of anti-HIV agents acting on different targets have been approved to combat HIV/AIDS. Among the abundant heterocyclic families, quinoline and isoquinoline moieties are recognized as promising scaffolds for HIV inhibition. This review intends to highlight the advances in diverse chemical structures and abundant biological activity of quinolines and isoquinolines as anti-HIV agents acting on different targets, which aims to provide useful references and inspirations to design and develop novel HIV inhibitors for medicinal chemists.


Subject(s)
Acquired Immunodeficiency Syndrome , Anti-HIV Agents , HIV Protease Inhibitors , HIV-1 , Quinolines , Humans , Saquinavir/therapeutic use , Acquired Immunodeficiency Syndrome/drug therapy , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Isoquinolines/pharmacology , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use
2.
Dis Colon Rectum ; 66(3): 360-365, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36599112

ABSTRACT

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 ).


Subject(s)
Anti-Infective Agents , Anus Neoplasms , Humans , Mice , Animals , Saquinavir/pharmacology , Saquinavir/therapeutic use , Human Papillomavirus Viruses , Protease Inhibitors , Mice, Transgenic , Anus Neoplasms/drug therapy , Anus Neoplasms/pathology , Retrospective Studies
3.
Antiviral Res ; 208: 105463, 2022 12.
Article in English | MEDLINE | ID: mdl-36372295

ABSTRACT

HIV-associated neurocognitive disorder (HAND) is prevalent in people living with HIV, despite the use of antiretroviral therapy (ART). Although several risk factors have been proposed to be related to HAND, substantial effort has been made to explore the neurotoxic effects of ART on HAND. HIV protease inhibitor (PI), an essential component of ART, has neurotoxicity in vivo and in vitro, which can contribute to the development of HAND. However, the pathogenesis of PI-associated neurotoxicity remains unclear. Here, we explored whether PI treatment is a potential pathogenic factor for HAND and elucidated its potential mechanisms. In our study, U87 cells were exposed to PIs, including lopinavir (LPV), ritonavir (RTV), darunavir, indinavir, and saquinavir at different concentrations, we found that LPV, LPV/RTV, and saquinavir attenuated autophagy in U87 cells, the results of Western blot showed that the expression of p62 dramatically was elevated and the level of LC3II/LC3I was decreased. Moreover, comparative transcriptomics revealed the involvement of the inflammatory response in the physiological activities of U87 cells exposed to LPV, with differential genes significantly enriched in the p38 MAPK signaling pathway. In the following study, we verified the results from RNA-sequence using the liquid chip technique, qRT-PCR, Elisa, and western blots, which suggested that LPV induced inflammatory response and the p38 MAPK pathway was involved in this process. Collectively, we demonstrated that PIs attenuated the involvement of astrocyte autophagy in inflammation via the p38 MAPK pathway, providing new insights into the mechanism of HAND.


Subject(s)
HIV Infections , HIV Protease Inhibitors , Humans , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , Saquinavir/pharmacology , Saquinavir/therapeutic use , p38 Mitogen-Activated Protein Kinases , Astrocytes , Ritonavir/pharmacology , HIV Infections/drug therapy , Inflammation/drug therapy , Autophagy , HIV Protease/therapeutic use
4.
Int J Mol Sci ; 23(20)2022 Oct 13.
Article in English | MEDLINE | ID: mdl-36293096

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Male , Humans , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Saquinavir/pharmacology , Saquinavir/therapeutic use , Prostate , Antiviral Agents/therapeutic use , Cell Line, Tumor , Drug Repositioning , Drug Synergism , Antineoplastic Agents/pharmacology , Lung Neoplasms/drug therapy
5.
Virology ; 576: 96-104, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36206607

ABSTRACT

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.


Subject(s)
HIV Infections , HIV Protease Inhibitors , Virus Diseases , Female , Mice , Humans , Animals , Saquinavir/pharmacology , Saquinavir/therapeutic use , HIV Protease Inhibitors/therapeutic use , RNA, Viral , Viral Load , Papillomaviridae/genetics , Enzyme Inhibitors , Anthracenes
6.
Plast Reconstr Surg ; 150(6): 1264e-1274e, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36112847

ABSTRACT

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.


Subject(s)
HIV Protease Inhibitors , Saquinavir , Mice , Animals , Saquinavir/pharmacology , Saquinavir/metabolism , Saquinavir/therapeutic use , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/metabolism , HIV Protease Inhibitors/therapeutic use , Toll-Like Receptor 4/physiology , Osteogenesis , Skull/injuries
7.
Biomolecules ; 12(7)2022 07 05.
Article in English | MEDLINE | ID: mdl-35883499

ABSTRACT

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.


Subject(s)
COVID-19 Drug Treatment , HIV Infections , HIV Protease Inhibitors , Neoplasms , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacology , HIV Protease Inhibitors/therapeutic use , Humans , Neoplasms/drug therapy , SARS-CoV-2 , Saquinavir/pharmacology , Saquinavir/therapeutic use
8.
Medicine (Baltimore) ; 100(31): e26787, 2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34397829

ABSTRACT

BACKGROUND: Lopinavir, ritonavir, atazanavir, and saquinavir had been reportedly used or suggested for coronavirus disease 2019 (COVID-19) treatment. They may cause electrocardiography changes. We aim to evaluate risk of PR prolongation, QRS widening, and QT prolongation from lopinavir, ritonavir, atazanavir, and saquinavir. METHODS: In accordance with preferred reporting items for systematic reviews and meta-analyses guidelines, our search was conducted in PubMed Central, PubMed, EBSCOhost, and ProQuest from inception to June 25, 2020. Titles and abstracts were reviewed for relevance. Cochrane Risk of Bias Tool 2.0 and Downs and Black criteria was used to evaluate quality of studies. RESULTS: We retrieved 9 articles. Most randomized controlled trials have low risk of biases while all quasi-experimental studies have a positive rating. Four studies reporting PR prolongation however only 2 studies with PR interval >200 ms. One of which, reported its association after treatment with ritonavir-boosted saquinavir treatment while another, during treatment with ritonavir-boosted atazanavir. No study reported QRS widening >120 ms with treatment. Four studies reporting QT prolongation, with only one study reaching QT interval >450 ms after ritonavir-boosted saquinavir treatment on healthy patients. There is only one study on COVID-19 patients reporting QT prolongation in 1 out of 95 patients after ritonavir-boosted lopinavir treatment. CONCLUSION: Limited evidence suggests that lopinavir, ritonavir, atazanavir, and saquinavir could cause PR prolongation, QRS widening, and QT prolongation. Further trials with closer monitoring and assessment of electrocardiography are needed to ascertain usage safety of antivirals in COVID-19 era.


Subject(s)
Atazanavir Sulfate/adverse effects , Long QT Syndrome/etiology , Lopinavir/adverse effects , Ritonavir/adverse effects , Saquinavir/adverse effects , Adult , Atazanavir Sulfate/therapeutic use , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Drug Therapy, Combination/methods , Drug Therapy, Combination/standards , Electrocardiography/methods , Humans , Lopinavir/therapeutic use , Ritonavir/therapeutic use , Saquinavir/therapeutic use
9.
Cell Death Dis ; 12(1): 67, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431821

ABSTRACT

Imbalance of macrophage polarization plays an indispensable role in acute lung injury (ALI), which is considered as a promising target. Matrix metalloproteinase-9 (MMP-9) is expressed in the macrophage, and has a pivotal role in secreting inflammatory cytokines. We reported that saquinavir (SQV), a first-generation human immunodeficiency virus-protease inhibitor, restricted exaggerated inflammatory response. However, whether MMP-9 could regulate macrophage polarization and inhibit by SQV is still unknown. We focused on the important role of macrophage polarization in CLP (cecal ligation puncture)-mediated ALI and determined the ability of SQV to maintain M2 over M1 phenotype partially through the inhibition of MMP-9. We also performed a limited clinical study to determine if MMP-9 is a biomarker of sepsis. Lipopolysaccharide (LPS) increased MMP-9 expression and recombinant MMP-9 (rMMP-9) exacerbated LPS-mediated M1 switching. Small interfering RNA to MMP-9 inhibited LPS-mediated M1 phenotype and SQV inhibition of this switching was reversed with rMMP-9, suggesting an important role for MMP-9 in mediating LPS-induced M1 phenotype. MMP-9 messenger RNA levels in peripheral blood mononuclear cells of these 14 patients correlated with their clinical assessment. There was a significant dose-dependent decrease in mortality and ALI after CLP with SQV. SQV significantly inhibited LPS-mediated M1 phenotype and increased M2 phenotype in cultured RAW 264.7 and primary murine bone marrow-derived macrophages as well as lung macrophages from CLP-treated mice. This study supports an important role for MMP-9 in macrophage phenotypic switching and suggests that SQV-mediated inhibition of MMP-9 may be involved in suppressing ALI during systemic sepsis.


Subject(s)
Acute Lung Injury/therapy , Macrophage Activation/physiology , Matrix Metalloproteinase 9/metabolism , Saquinavir/therapeutic use , Animals , Disease Models, Animal , HIV Protease Inhibitors/therapeutic use , Humans , Male , Mice , Middle Aged
10.
Travel Med Infect Dis ; 35: 101646, 2020.
Article in English | MEDLINE | ID: mdl-32294562

ABSTRACT

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.


Subject(s)
Betacoronavirus/chemistry , Coronavirus Infections/virology , Cysteine Endopeptidases/chemistry , Drug Discovery/methods , Pneumonia, Viral/virology , Spike Glycoprotein, Coronavirus/chemistry , Viral Nonstructural Proteins/chemistry , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/therapeutic use , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/therapeutic use , Binding Sites , COVID-19 , Coronavirus 3C Proteases , Coronavirus Infections/drug therapy , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/therapeutic use , Humans , Indinavir/chemistry , Indinavir/therapeutic use , Molecular Docking Simulation , Off-Label Use , Pandemics , Pneumonia, Viral/drug therapy , SARS-CoV-2 , Saquinavir/chemistry , Saquinavir/therapeutic use , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Structural Homology, Protein , Viral Nonstructural Proteins/antagonists & inhibitors , Zanamivir/chemistry , Zanamivir/therapeutic use , COVID-19 Drug Treatment
11.
Mol Med ; 23: 92-100, 2017 06.
Article in English | MEDLINE | ID: mdl-28332696

ABSTRACT

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.


Subject(s)
Carotid Artery Injuries/metabolism , Cathepsin L/metabolism , Hyperplasia/metabolism , Tunica Intima/pathology , Animals , Carotid Artery Injuries/drug therapy , Cathepsin L/genetics , Cells, Cultured , HIV Protease Inhibitors/therapeutic use , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Hyperplasia/drug therapy , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Saquinavir/therapeutic use , Toll-Like Receptor 4/metabolism
12.
Int J Cancer ; 140(8): 1713-1726, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27870005

ABSTRACT

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.


Subject(s)
HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , Neoplasms/drug therapy , Sarcoma, Kaposi/drug therapy , Antiretroviral Therapy, Highly Active , HIV Infections/complications , HIV Infections/virology , HIV-1/drug effects , HIV-1/pathogenicity , Humans , Neoplasms/complications , Neoplasms/virology , Nitric Oxide/metabolism , Saquinavir/analogs & derivatives , Saquinavir/therapeutic use , Sarcoma, Kaposi/complications , Sarcoma, Kaposi/virology
13.
Mediators Inflamm ; 2017: 7083528, 2017.
Article in English | MEDLINE | ID: mdl-29440779

ABSTRACT

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.


Subject(s)
HMGB1 Protein/physiology , JNK Mitogen-Activated Protein Kinases/physiology , Liver/blood supply , Lung Injury/drug therapy , Reperfusion Injury/drug therapy , Saquinavir/therapeutic use , Signal Transduction/physiology , Toll-Like Receptor 4/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cytokines/physiology , Lung Injury/immunology , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/immunology , Warm Ischemia
16.
AIDS ; 27(10): 1674-7, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23907270

ABSTRACT

HIV and malaria overlap geographically, but the full impact of different antiretrovirals on malaria remains poorly understood. We examined the antimalarial activity of the HIV protease inhibitors lopinavir and saquinavir and the non-nucleoside reverse transcriptase inhibitor nevirapine on Plasmodium falciparum liver stages. Our results demonstrate that the HIV PI lopinavir inhibits liver stage parasites at clinically relevant concentrations, that is, at drug levels achieved in HIV-infected patients on standard dosing regimens. Because drugs that inhibit liver stages target parasites when they are present in lower numbers, these results might have implications for eradication efforts.


Subject(s)
HIV Protease Inhibitors/pharmacology , Liver/parasitology , Plasmodium falciparum/drug effects , Reverse Transcriptase Inhibitors/pharmacology , HIV Infections/complications , HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , Humans , In Vitro Techniques , Liver/drug effects , Lopinavir/pharmacology , Lopinavir/therapeutic use , Malaria, Falciparum/complications , Nevirapine/pharmacology , Nevirapine/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Saquinavir/pharmacology , Saquinavir/therapeutic use
17.
J Exp Clin Cancer Res ; 32: 38, 2013 Jun 08.
Article in English | MEDLINE | ID: mdl-23759068

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Leukemic/drug effects , HIV Protease Inhibitors/pharmacology , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Saquinavir/pharmacology , Telomerase/genetics , Telomerase/metabolism , Catalytic Domain/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , HIV Protease Inhibitors/therapeutic use , Humans , Jurkat Cells , Leukemia, T-Cell/drug therapy , Proto-Oncogene Proteins c-myc/metabolism , Saquinavir/therapeutic use , Telomerase/chemistry , Transcription, Genetic/drug effects
18.
Ter Arkh ; 85(11): 16-20, 2013.
Article in Russian | MEDLINE | ID: mdl-24432594

ABSTRACT

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.


Subject(s)
HIV Infections/drug therapy , HIV , Perinatal Care/methods , Pregnancy Complications, Infectious , Saquinavir/therapeutic use , Antiretroviral Therapy, Highly Active/methods , Female , Follow-Up Studies , HIV Infections/epidemiology , HIV Infections/transmission , HIV Protease Inhibitors/therapeutic use , Humans , Incidence , Pregnancy , Retrospective Studies , Russia/epidemiology , Treatment Outcome
19.
PLoS One ; 7(9): e44575, 2012.
Article in English | MEDLINE | ID: mdl-22984529

ABSTRACT

OBJECTIVE: In a nationwide, population-based cohort study we assessed the risk of diabetes mellitus (DM) in HIV-infected individuals compared with the general population, and evaluated the impact of risk factors for DM in HIV-infected individuals. METHODS: We identified 4,984 Danish-born HIV-infected individuals from the Danish HIV Cohort Study and a Danish born population-based age- and gender-matched comparison cohort of 19,936 individuals (study period: 1996-2009). Data on DM was obtained from the Danish National Hospital Registry and the Danish National Prescription Registry. Incidence rate ratios (IRR) and impact of risk factors including exposure to Highly Active Antiretroviral Therapy (HAART) and antiretroviral drugs were estimated by Poisson regression analyses. RESULTS: In the period 1996-1999 risk of DM was higher in HIV-infected individuals compared to the comparison cohort (adjusted IRR: 2.83; 95%CI: 1.57-5.09), both before (adjusted IRR: 2.40; 95%CI: 1.03-5.62) and after HAART initiation (adjusted IRR: 3.24; 95% CI: 1.42-7.39). In the period 1999-2010 the risk of DM in HIV-infected individuals did not differ from that of the comparison cohort (adjusted IRR: 0.90; 95% CI: 0.72-1.13), although the risk was decreased before HAART-initiation (adjusted IRR: 0.45; 95%CI: 0.21-0.96). Increasing age, BMI and the presence of lipoatrophy increased the risk of DM, as did exposure to indinavir, saquinavir, stavudine and didanosine. CONCLUSION: Native HIV-infected individuals do not have an increased risk of developing DM compared to a native background population after year 1998. Some antiretroviral drugs, not used in modern antiretroviral treatment, seem to increase the risk of DM.


Subject(s)
Diabetes Mellitus/diagnosis , HIV Infections/complications , Adult , Anti-Retroviral Agents/therapeutic use , Antiretroviral Therapy, Highly Active/methods , Body Mass Index , Cohort Studies , Denmark , Diabetes Complications/diagnosis , Didanosine/therapeutic use , Female , HIV Infections/diagnosis , HIV Infections/therapy , Humans , Incidence , Indinavir/therapeutic use , Male , Middle Aged , Regression Analysis , Risk Factors , Saquinavir/therapeutic use , Stavudine/therapeutic use , Treatment Outcome
20.
Med Mal Infect ; 42(9): 421-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22938775

ABSTRACT

OBJECTIVE: The authors had for aim to describe the effectiveness and the safety of a saquinavir/ritonavir (SQV/r) regimen, 1000/100mg twice daily, in HIV-infected pregnant patients. PATIENTS AND METHOD: We made a prospective and observational study of HIV positive female patients beginning or going on SQV/r antiretroviral treatment (ART) during pregnancy. RESULTS: Sixty-two patients were enrolled from July 2007 to June 2009 in 10 infectious diseases units in France. Thirty-six women (group 1) were ART naive on inclusion, 20 (group 2) had been previously treated and then switched to SQV/r, six (group 3) were treated with SQV/r before pregnancy. 58 patients delivered while on SQV/r regimen after a median pregnancy duration of 39 WA. Eighty percent had a viral load below 50 copies/mL and 93% below 400 copies/mL: respectively 77% and 93.5% in group 1, 83% and 89% in group 2, 83% and 100% in group 3. The median SQV minimum concentrations (C(min)) measured at the third trimester and at delivery were adequate, respectively 0.91 mg/L and 0.86 mg/L. Most women (52%) had a vaginal delivery; 12 (21%) had an elective caesarean section, for obstetrics factors in eight cases. None of the newborns were HIV-infected at 6 months of age (n = 59, one death at day 3). Only one severe adverse event occurred due to saquinavir (maternal grade 3 hepatotoxicity). CONCLUSION: SQV/r 1000/100mg twice daily seems to be effective and safe in HIV-infected pregnant women with adequate saquinavir C(min).


Subject(s)
HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , HIV-1 , HIV-2 , Pregnancy Complications, Infectious/drug therapy , Ritonavir/therapeutic use , Saquinavir/therapeutic use , Adult , CD4 Lymphocyte Count , Chemical and Drug Induced Liver Injury/etiology , Cohort Studies , Delivery, Obstetric/statistics & numerical data , Drug Therapy, Combination , Female , HIV Infections/congenital , HIV Infections/prevention & control , HIV Infections/transmission , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/adverse effects , Humans , Infant, Newborn , Infectious Disease Transmission, Vertical/prevention & control , Male , Pregnancy , Pregnancy Outcome , Prospective Studies , Ritonavir/administration & dosage , Ritonavir/adverse effects , Saquinavir/administration & dosage , Saquinavir/adverse effects , Treatment Outcome , Viral Load , Viremia/drug therapy , Young Adult , Zidovudine/administration & dosage , Zidovudine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL