Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Vaccines (Basel) ; 12(2)2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38400152

ABSTRACT

The development of mRNA vaccines has increased rapidly since the COVID-19 pandemic. As one of the critical attributes, understanding mRNA lipid nanoparticle (LNP) stability is critical in the vaccine product development. However, the correlation between LNPs' physiochemical characteristics and their potency still remains unclear. The lack of regulatory guidance on the specifications for mRNA LNPs is also partially due to this underexplored relationship. In this study, we performed a three-month stability study of heat-stressed mRNA LNP samples. The mRNA LNP samples were analyzed for their mRNA degradation, LNP particle sizes, and mRNA encapsulation efficiency. In vitro cell potency was also evaluated and correlated with these above-mentioned physiochemical characterizations. The mRNA degradation-cell potency correlation data showed two distinct regions, indicating a critical cut-off size limit for mRNA degradation. The same temperature dependence was also observed in the LNP size-cell potency correlation.

2.
J Pharm Biomed Anal ; 234: 115523, 2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37336039

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of severe lower respiratory tract infections worldwide. A safe and effective RSV vaccine has been an elusive goal but recent advances in vaccine technology have improved the likelihood that a vaccine for the prevention of RSV could be licensed in near future. We have developed an RSV vaccine V171 consisting of four lipids and messenger ribonucleic acid (mRNA) encoding an engineered form of the RSV F protein stabilized in its prefusion conformation. The lipids form lipid nanoparticles (LNP) with mRNA encapsulated during process, which protects the mRNA from degradation and enables the mRNA to be delivered into mammalian cells. Once inside the cells, the mRNA then can be translated into RSV F protein and elicit both humoral and cellular immune responses. Preclinical results and Phase I clinical trial results indicate that this mRNA vaccine targeting RSV F protein is a promising RSV vaccine approach and should be further evaluated in clinical trials. We have developed a cell-based relative potency assay to support the Phase II development of this vaccine. Test articles and a reference standard are tested with serial dilutions in a 96-well plate pre-seeded with Hep G2 cells. Cells were incubated for 16-18 h after transfection and then permeabilized and stained with a human monoclonal antibody specific to RSV F protein, followed by a fluorophore-conjugated secondary antibody. The plate is then analyzed for percentage of transfected cells and relative potency of the test article is calculated by comparing its EC50 to that of a reference standard. This assay takes advantage of the fact that due to the inherent variability in biological test systems an absolute measure of potency is more variable than a measure of activity relative to a standard. Targeting testing relative potency range 25-250 %, our assay showed an R2 close to 1 for linearity, relative bias of 1.05-5.41 %, and intermediate precision of 11.0 %. The assay has been used for testing of process development samples, formulation development samples, as well as drug product intermediate (DPI) and drug product (DP) in support of Phase II development of our RSV mRNA vaccine.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Animals , Humans , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus Infections/prevention & control , Antibodies, Neutralizing , Antibodies, Viral , Respiratory Syncytial Virus Vaccines/genetics , RNA, Messenger/genetics , Lipids , Mammals/genetics , mRNA Vaccines
3.
Vaccine ; 33(1): 252-9, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-24951860

ABSTRACT

The toxicity of Clostridium difficile large clostridial toxin B (TcdB) can be reduced by many orders of magnitude by a combination of targeted point mutations. However, a TcdB mutant with five point mutations (referred to herein as mTcdB) still has residual toxicity that can be detected in cell-based assays and in-vivo mouse toxicity assays. This residual toxicity can be effectively removed by treatment with formaldehyde in solution. Storage of the formaldehyde-treated mTcdB as a liquid can result in reversion over time back to the mTcdB level of toxicity, with the rate of reversion dependent on the storage temperature. We found that for both the "forward" mTcdB detoxification reaction with formaldehyde, and the "reverse" reversion to toxicity reaction, mouse toxicity correlated with several biochemical assays including anion exchange chromatography retention time and appearance on SDS-PAGE. Maintenance of a low concentration of formaldehyde prevents reversion to toxicity in liquid formulations. However, when samples with 0.016% (v/v) formaldehyde were lyophilized and stored at 37 °C, formaldehyde continued to react with and modify the mTcdB in the lyophilized state. Lyophilization alone effectively prevented reversion to toxicity for formaldehyde-treated, formaldehyde-removed mTcdB samples stored at 37 °C for 6 months. Formaldehyde-treated, formaldehyde-removed lyophilized mTcdB showed no evidence of reversion to toxicity, appeared stable by several assays, and was immunogenic in mice, even after storage for 6 months at 37 °C.


Subject(s)
Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Bacterial Vaccines/toxicity , Formaldehyde/metabolism , Toxoids/toxicity , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Toxins/chemistry , Bacterial Toxins/immunology , Bacterial Vaccines/chemistry , Bacterial Vaccines/immunology , Bacterial Vaccines/radiation effects , Chromatography, Ion Exchange , Drug Storage , Electrophoresis, Polyacrylamide Gel , Female , Freeze Drying , Mice, Inbred C57BL , Mutant Proteins/chemistry , Mutant Proteins/immunology , Mutant Proteins/toxicity , Temperature , Time Factors , Toxoids/chemistry , Toxoids/immunology
4.
J Pharmacol Toxicol Methods ; 63(3): 227-35, 2011.
Article in English | MEDLINE | ID: mdl-21147239

ABSTRACT

INTRODUCTION: Pharmacokinetic properties of biotherapeutics are an important aspect of preclinical drug development. The lead identification and optimization space is characterized by aggressive timelines, large sample numbers, a variety of species and matrices, and limited reagent and sample volumes all of which represent challenges for traditional microtiter plate assays. Since the Gyrolab immunoassay platform can accommodate small sample volumes and automated assay processing, we evaluated the workstation as an alternative to the plate-based assays. METHODS: Three representative example assays--a generic anti-human IgG, a target specific and an anti-drug capture assay--were investigated in detail for accuracy and precision performance and their application to bioanalytical support for preclinical pharmacokinetic studies. Different animal matrices were tested in the assays and during study support. RESULTS: Gyrolab procedures could be closely modeled after regular microtiter plate assays. The small reagent volumes necessary for Gyrolab allowed studying serial bleeds of transgenic mice with only 10µL of blood sample. During development and during study support, the Gyrolab performance was similar to what can be expected from plate-based systems with accuracy and precision within 100 ± 20% or less. DISCUSSION: Overall, the technology was well suited to support quantitation of biotherapeutics using small volume samples from different preclinical species. Limited operator involvement for assay processing allowed for reduced staffing and training. However, high instrument costs and a single source of reagent supplies represent risks when moving assays further into long-term applications such as clinical studies. Despite interest in the bioanalytical field, this is the first detailed investigation of bioanalytical applications of Gyrolab in pharmacokinetic studies.


Subject(s)
Biopharmaceutics/methods , Drug Evaluation, Preclinical/methods , Immunoassay/methods , Pharmacokinetics , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Biopharmaceutics/instrumentation , Drug Evaluation, Preclinical/instrumentation , Enzyme-Linked Immunosorbent Assay , Humans , Immunoassay/instrumentation , Macaca fascicularis , Macaca mulatta , Mice , Mice, Transgenic , Miniaturization , Rats , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Reproducibility of Results , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/pharmacokinetics
5.
Virology ; 353(2): 482-90, 2006 Sep 30.
Article in English | MEDLINE | ID: mdl-16859727

ABSTRACT

In the current study, we extended our previous works on natural endogenous reverse transcription (NERT) and further examined its potential as a virucide molecular target in sexual transmission of primate lentiviruses. HIV-1 and SIV virions were pretreated with select nucleoside (NRTIs) and nonnucleoside RT inhibitors (NNRTIs), either alone or in combination with NERT-stimulating substances. The effects of these antiretrovirals on virion inactivation were analyzed in human T cell lines and primary cell cultures. Pretreatment of HIV-1 virions with physiologic NERT-stimulants and 3'-azido-3'-deoxythymidine 5'-triphosphate (AZT-TP) or nevirapine potently inactivated cell-free HIV-1 virions and resulted in strong inhibition of the viral infectivity. Pretreatment of chimeric SHIV-RT virions with NERT-stimulating cocktail and select antiretrovirals also resulted in virion inactivation and inhibition of viral infectivity in T cell lines. Our findings demonstrate the potential clinical utility of approaches based on inhibiting NERT in sexual transmission of HIV-1, through the development of effective anti-HIV-1 microbicides, such as NRTIs and NNRTIs.


Subject(s)
Antiviral Agents/pharmacology , HIV-1/drug effects , Nevirapine/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Simian Immunodeficiency Virus/drug effects , Thymine Nucleotides/pharmacology , Transcription, Genetic/drug effects , Zidovudine/analogs & derivatives , Cells, Cultured , Dideoxynucleotides , Disease Transmission, Infectious/prevention & control , HIV-1/genetics , Humans , Lentivirus Infections/prevention & control , Lentivirus Infections/transmission , Simian Immunodeficiency Virus/genetics , T-Lymphocytes , Zidovudine/pharmacology
6.
J Virol ; 78(19): 10433-41, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367609

ABSTRACT

A novel entry mechanism has been proposed for the avian sarcoma and leukosis virus (ASLV), whereby interaction with specific cell surface receptors activates or primes the viral envelope glycoprotein (Env), rendering it sensitive to subsequent low-pH-dependent fusion triggering in acidic intracellular organelles. However, ASLV fusion seems to proceed to a lipid mixing stage at neutral pH, leading to the suggestion that low pH might instead be required for a later stage of viral entry such as uncoating (L. J. Earp, S. E. Delos, R. C. Netter, P. Bates, and J. M. White. J. Virol. 77:3058-3066, 2003). To address this possibility, hybrid virus particles were generated with the core of human immunodeficiency virus type 1 (HIV-1), a known pH-independent virus, and with subgroups A or B ASLV Env proteins. Infection of cells by these pseudotyped virions was blocked by lysosomotropic agents, as judged by inhibition of HIV-1 DNA synthesis. Furthermore, by using HIV-1 cores that contain a Vpr-beta-lactamase fusion protein (Vpr-BlaM) to monitor viral penetration into the cytosol, we demonstrated that virions bearing ASLV Env, but not HIV-1 Env, enter the cytosol in a low-pH-dependent manner. This effect was independent of the presence of the cytoplasmic tail of ASLV Env. These studies provide strong support for the model, indicating that low pH is required for ASLV Env-dependent viral penetration into the cytosol and not for viral uncoating.


Subject(s)
Avian Leukosis Virus/physiology , Avian Sarcoma Viruses/physiology , Gene Products, env/metabolism , Avian Leukosis Virus/genetics , Avian Leukosis Virus/growth & development , Avian Sarcoma Viruses/genetics , Avian Sarcoma Viruses/growth & development , Biological Transport , Cell Line , Cytosol/virology , DNA, Viral/biosynthesis , Gene Products, env/genetics , Genes, Reporter , HIV-1/genetics , HIV-1/growth & development , Humans , Hydrogen-Ion Concentration , Organelles/metabolism , Organelles/virology , Receptors, Virus/physiology , Recombination, Genetic , beta-Lactamases/genetics , beta-Lactamases/metabolism
7.
Science ; 305(5683): 528-32, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15247437

ABSTRACT

We describe the efficacy of L-870812, an inhibitor of HIV-1 and SIV integrase, in rhesus macaques infected with the simian-human immunodeficiency virus (SHIV) 89.6P. When initiated before CD4 cell depletion, L-870812 therapy mediated a sustained suppression of viremia, preserving CD4 levels and permitting the induction of virus-specific cellular immunity. L-870812 was also active in chronic infection; however, the magnitude and durability of the effect varied in conjunction with the pretreatment immune response and viral load. These studies demonstrate integrase inhibitor activity in vivo and suggest that cellular immunity facilitates chemotherapeutic efficacy in retroviral infections.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Acquired Immunodeficiency Syndrome/immunology , HIV-1/physiology , Integrase Inhibitors/therapeutic use , Naphthyridines/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology , Acquired Immunodeficiency Syndrome/virology , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/blood , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Drug Resistance, Viral , HIV Integrase/genetics , HIV Integrase/metabolism , HIV Integrase Inhibitors/administration & dosage , HIV Integrase Inhibitors/blood , HIV Integrase Inhibitors/pharmacology , HIV Integrase Inhibitors/therapeutic use , HIV-1/drug effects , HIV-1/enzymology , HIV-1/genetics , Immunity, Cellular , Integrase Inhibitors/administration & dosage , Integrase Inhibitors/blood , Integrase Inhibitors/pharmacology , Integrases/genetics , Integrases/metabolism , Leukocytes, Mononuclear/virology , Macaca mulatta , Mutation , Naphthyridines/administration & dosage , Naphthyridines/blood , Naphthyridines/pharmacology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/enzymology , Simian Immunodeficiency Virus/genetics , Viral Load , Viremia/drug therapy , Virus Replication/drug effects
8.
J Virol ; 77(3): 2271-5, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12525664

ABSTRACT

Three of five virally suppressed human immunodeficiency virus type I (HIV-1)-infected patients treated with highly active antiretroviral therapy and followed intensively with a supersensitive reverse transcriptase PCR assay with a lower limit of quantitation of 5 copies/ml showed statistically significant viral load decays below 50 copies/ml, with half-lives of 5 to 8 months and a mean of 6 months. This range of half-lives is consistent with the estimated half-life of the latent HIV-1 reservoir in the peripheral blood. Those patients without decay of viral load in plasma may have significant cryptic HIV-1 residual replication.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Antiretroviral Therapy, Highly Active , HIV-1/drug effects , RNA, Viral/blood , Acquired Immunodeficiency Syndrome/virology , CD4-Positive T-Lymphocytes/virology , Half-Life , Humans , Regression Analysis , Reverse Transcriptase Polymerase Chain Reaction , Viral Load
9.
AIDS Res Hum Retroviruses ; 19(12): 1097-103, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14709246

ABSTRACT

Dendritic cells (DCs) are potent antigen-presenting cells, and their physiological localization in tissues that interact with the external environment is important as a first barrier against pathogens such as human immunodeficiency virus type I (HIV-1). Several models have been proposed to explain the possible role of DCs as a reservoir for HIV-1 in patients on virally suppressive highly active antiretroviral therapy (HAART). However, the low yield of cell isolates has made this evaluation a difficult task. The present study analyzes whether peripheral blood DCs from HIV-1-infected individuals on virally suppressive HAART, with plasma HIV-1 RNA levels of less than 50 copies/ml, carry either HIV-1 provirus and/or HIV-1 virions. Peripheral blood DCs were isolated from a cohort of 10 HIV-1-seropositive men taking suppressive HAART. In five patients, plasmacytoid and myeloid dendritic cells were isolated to attempt to identify their respective roles in HIV-1 residual disease. Viral out-growth assays were performed in vitro, as well as gag and R/U5 polymerase chain reaction (PCR) amplification of viral RNA and DNA, respectively, from DC and peripheral blood mononuclear cell (PBMC) extracts. Fluorescence activated cell-sorting (FACS) data revealed cellular yields from 85.90 to 95.18%, of relatively pure DCs isolated from patients' PBMCs. Although HIV-1 RNA gag and DNA RU/5 were detected in all PBMC samples isolated from the patients, proviral DNA and viral RNA forms were not detected in any of the DC isolates. In addition, no replication-competent virus was demonstrated in DC coculture assays, while virus was isolated from each patients' CD8+ T-lymphocyte-depleted PBMC cocultures. Furthermore, HIV-1 gag proviral DNA was not detected in either plasmacytoid or myeloid DC subfractions. The current study suggests that in HIV-1-infected individuals treated with suppressive HAART, peripheral blood DCs do not carry HIV-1 proviral DNA or viral particles attached to their surface. These populations of peripheral blood DCs are likely not a major HIV-1 reservoir in patients on HAART with clinically undetectable plasma viral RNA.


Subject(s)
Antiretroviral Therapy, Highly Active , Dendritic Cells/virology , HIV Infections/virology , HIV-1/physiology , Anti-HIV Agents/therapeutic use , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cohort Studies , DNA, Viral/blood , HIV Infections/blood , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/drug effects , HIV-1/isolation & purification , Humans , Leukocytes, Mononuclear/virology
10.
AIDS ; 16(1): 39-45, 2002 Jan 04.
Article in English | MEDLINE | ID: mdl-11741161

ABSTRACT

BACKGROUND: HIV-1-infected men on suppressive highly active antiretroviral therapy (HAART) have a reduction of viral replication in vivo, but HIV-1 RNA is still detectable by certain ultrasensitive reverse transcriptase-PCR assays in blood plasma. Replication-competent virus can also be isolated from both peripheral blood mononuclear cells (PBMC) and seminal cells of these patients. Despite HAART, on-going in vivo infection of HIV-1-seropositive patients' PBMC was demonstrated by the detection of episomal HIV-1 moieties, known as HIV-1 two-long terminal repeat (2-LTR) DNA circles. METHODS: The present study analyzes whether new cellular infections occur in vivo in seminal cells of HIV-1-infected men on suppressive HAART. PBMC and seminal cells were isolated from a cohort of HIV-1-seropositive men taking suppressive HAART (< 50 copies HIV RNA/ml blood plasma). Viral growth assays were performed in vitro, as well as semi-quantitative PCR to detect HIV-1 2-LTR circular DNA in PBMC and seminal mononuclear cells. RESULTS: Viral growth in vitro was demonstrated in 16 out of 28 (57%) patients' PBMC, and in five patients' seminal cells (18%). Although 18 patients' PBMC were positive for HIV-1 2-LTR DNA circles, importantly, 2-LTR circular DNA was not detected in any semen sample, even when replication-competent HIV-1 virus had been recovered from a patient's seminal cells by viral co-culture assays. CONCLUSIONS: The current study suggests that in HIV-1-infected men treated with suppressive HAART, new cellular infections occur in PBMC, but that new infections do not take place in seminal cells in vivo. Thus, these findings suggest that mainly latent HIV-1 occurs in seminal cells of men on suppressive HAART, which may be a compartment-specific mechanism of residual HIV-1 disease.


Subject(s)
Antiretroviral Therapy, Highly Active , HIV Infections/drug therapy , HIV-1/isolation & purification , Semen/virology , Virus Latency , DNA, Viral/analysis , DNA, Viral/blood , HIV Infections/immunology , HIV Infections/virology , HIV Long Terminal Repeat/genetics , HIV-1/drug effects , HIV-1/physiology , Humans , Leukocytes, Mononuclear/virology , Male , Polymerase Chain Reaction , RNA, Viral/blood , Semen/cytology , Semen/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...