Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Med Chem ; 67(2): 1421-1446, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38190324

ABSTRACT

Approved therapies for hepatitis B virus (HBV) treatment include nucleos(t)ides and interferon alpha (IFN-α) which effectively suppress viral replication, but they rarely lead to cure. Expression of viral proteins, especially surface antigen of the hepatitis B virus (HBsAg) from covalently closed circular DNA (cccDNA) and the integrated genome, is believed to contribute to the persistence of HBV. This work focuses on therapies that target the expression of HBV proteins, in particular HBsAg, which differs from current treatments. Here we describe the identification of AB-452, a dihydroquinolizinone (DHQ) analogue. AB-452 is a potent HBV RNA destabilizer by inhibiting PAPD5/7 proteins in vitro with good in vivo efficacy in a chronic HBV mouse model. AB-452 showed acceptable tolerability in 28-day rat and dog toxicity studies, and a high degree of oral exposure in multiple species. Based on its in vitro and in vivo profiles, AB-452 was identified as a clinical development candidate.


Subject(s)
Hepatitis B virus , Hepatitis B, Chronic , Mice , Rats , Animals , Dogs , Hepatitis B virus/genetics , Hepatitis B Surface Antigens , Antiviral Agents/therapeutic use , Hepatitis B, Chronic/drug therapy , RNA, Viral/genetics , Structure-Activity Relationship , Naphthyridines/pharmacology , Naphthyridines/therapeutic use , DNA, Viral/genetics , Virus Replication
2.
Bioorg Med Chem Lett ; 94: 129456, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37633618

ABSTRACT

Disruption of the HBV capsid assembly process through small-molecule interaction with HBV core protein is a validated target for the suppression of hepatitis B viral replication and the development of new antivirals. Through combination of key structural features associated with two distinct series of capsid assembly modulators, a novel aminochroman-based chemotype was identified. Optimization of anti-HBV potency through generation of SAR in addition to further core modifications provided a series of related functionalized aminoindanes. Key compounds demonstrated excellent cellular potency in addition to favorable ADME and pharmacokinetic profiles and were shown to be highly efficacious in a mouse model of HBV replication. Aminoindane derivative AB-506 was subsequently advanced into clinical development.


Subject(s)
Antiviral Agents , Capsid Proteins , Capsid , Animals , Mice , Antiviral Agents/pharmacology , Disease Models, Animal , Structure-Activity Relationship , Hepatitis B virus/drug effects , Hepatitis B virus/metabolism
3.
Hepatol Commun ; 6(12): 3457-3472, 2022 12.
Article in English | MEDLINE | ID: mdl-36194181

ABSTRACT

AB-506 is a potent, pan-genotypic small molecule capsid inhibitor that inhibits hepatitis B virus (HBV) pregenomic RNA encapsidation. We assessed the safety, pharmacokinetics, and antiviral activity of AB-506 in two randomized, double-blinded Phase 1 studies in healthy subjects (HS) and subjects with chronic HBV infection (CHB). Single ascending and multiple doses of AB-506 or placebo (30-1000 mg or 400 mg daily for 10 days) were assessed in HS. AB-506 or placebo was assessed at either 160 mg or 400 mg daily for 28 days in subjects with CHB. A second follow-up study examined AB-506 or placebo at 400 mg daily for 28 days in 14 Caucasian and 14 East-Asian HS. Twenty-eight days of AB-506 at 160 mg and 400 mg produced mean HBV-DNA declines from baseline of 2.1 log10 IU/ml and 2.8 log10 IU/ml, respectively. Four subjects with CHB (all Asian) had Grade 4 alanine aminotransferase (ALT) elevations (2 at each dose) as HBV DNA was declining; three events led to treatment discontinuation. In the second follow-up study, 2 Asian HS had serious transaminitis events leading to treatment and study termination. No subjects had bilirubin elevations or signs of hepatic decompensation. Conclusion: AB-506 demonstrated mean HBV-DNA declines of >2 log10 ; however, transient but severe ALT flares were observed in 4 Asian subjects with CHB. In the follow-up study in HS, 2 additional Asian HS had Grade 4 flares, suggesting that AB-506 hepatotoxicity contributed to the ALT elevations. The AB-506 development program was terminated because of these findings.


Subject(s)
Antiviral Agents , Hepatitis B , Humans , Antiviral Agents/adverse effects , Capsid , Capsid Proteins , DNA, Viral , Follow-Up Studies , Healthy Volunteers , Hepatitis B/drug therapy , Hepatitis B e Antigens , Hepatitis B virus/genetics
4.
Antiviral Res ; 197: 105211, 2022 01.
Article in English | MEDLINE | ID: mdl-34826506

ABSTRACT

AB-506, a small-molecule inhibitor targeting the HBV core protein, inhibits viral replication in vitro (HepAD38 cells: EC50 of 0.077 µM, CC50 > 25 µM) and in vivo (HBV mouse model: ∼3.0 log10 reductions in serum HBV DNA compared to the vehicle control). Binding of AB-506 to HBV core protein accelerates capsid assembly and inhibits HBV pgRNA encapsidation. Furthermore, AB-506 blocks cccDNA establishment in HBV-infected HepG2-hNTCP-C4 cells and primary human hepatocytes, leading to inhibition of viral RNA, HBsAg, and HBeAg production (EC50 from 0.64 µM to 1.92 µM). AB-506 demonstrated activity across HBV genotypes A-H and maintains antiviral activity against nucleos(t)ide analog-resistant variants in vitro. Evaluation of AB-506 against a panel of core variants showed that T33N/Q substitutions results in >200-fold increase in EC50 values, while L30F, L37Q, and I105T substitutions showed an 8 to 20-fold increase in EC50 values in comparison to the wild-type. In vitro combinations of AB-506 with NAs or an RNAi agent were additive to moderately synergistic. AB-506 exhibits good oral bioavailability, systemic exposure, and higher liver to plasma ratios in rodents, a pharmacokinetic profile supporting clinical development for chronic hepatitis B.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B virus/drug effects , Viral Core Proteins/antagonists & inhibitors , Virus Replication/drug effects , Animals , Antiviral Agents/pharmacokinetics , Cells, Cultured , Drug Evaluation, Preclinical , Female , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Mice , Rats , Virus Assembly/drug effects
5.
J Virol ; 95(18): e0057421, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34191584

ABSTRACT

Noncanonical poly(A) polymerases PAPD5 and PAPD7 (PAPD5/7) stabilize hepatitis B virus (HBV) RNA via the interaction with the viral posttranscriptional regulatory element (PRE), representing new antiviral targets to control HBV RNA metabolism, hepatitis B surface antigen (HBsAg) production, and viral replication. Inhibitors targeting these proteins are being developed as antiviral therapies; therefore, it is important to understand how PAPD5/7 coordinate to stabilize HBV RNA. Here, we utilized a potent small-molecule AB-452 as a chemical probe, along with genetic analyses to dissect the individual roles of PAPD5/7 in HBV RNA stability. AB-452 inhibits PAPD5/7 enzymatic activities and reduces HBsAg both in vitro (50% effective concentration [EC50] ranged from 1.4 to 6.8 nM) and in vivo by 0.94 log10. Our genetic studies demonstrate that the stem-loop alpha sequence within PRE is essential for both maintaining HBV poly(A) tail integrity and determining sensitivity toward the inhibitory effect of AB-452. Although neither single knockout (KO) of PAPD5 nor PAPD7 reduces HBsAg RNA and protein production, PAPD5 KO does impair poly(A) tail integrity and confers partial resistance to AB-452. In contrast, PAPD7 KO did not result in any measurable changes within the HBV poly(A) tails, but cells with both PAPD5 and PAPD7 KO show reduced HBsAg production and conferred complete resistance to AB-452 treatment. Our results indicate that PAPD5 plays a dominant role in stabilizing viral RNA by protecting the integrity of its poly(A) tail, while PAPD7 serves as a second line of protection. These findings inform PAPD5-targeted therapeutic strategies and open avenues for further investigating PAPD5/7 in HBV replication. IMPORTANCE Chronic hepatitis B affects more than 250 million patients and is a major public health concern worldwide. HBsAg plays a central role in maintaining HBV persistence, and as such, therapies that aim at reducing HBsAg through destabilizing or degrading HBV RNA have been extensively investigated. Besides directly degrading HBV transcripts through antisense oligonucleotides or RNA silencing technologies, small-molecule compounds targeting host factors such as the noncanonical poly(A) polymerase PAPD5 and PAPD7 have been reported to interfere with HBV RNA metabolism. Herein, our antiviral and genetic studies using relevant HBV infection and replication models further characterize the interplays between the cis element within the viral sequence and the trans elements from the host factors. PAPD5/7-targeting inhibitors, with oral bioavailability, thus represent an opportunity to reduce HBsAg through destabilizing HBV RNA.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Directed DNA Polymerase/metabolism , Hepatitis B virus/genetics , Hepatitis B/virology , RNA Nucleotidyltransferases/metabolism , RNA Stability , RNA, Viral/chemistry , Virus Replication , Animals , Antiviral Agents/pharmacology , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/genetics , DNA-Directed DNA Polymerase/genetics , Enzyme Inhibitors/pharmacology , Hep G2 Cells , Hepatitis B/genetics , Hepatitis B/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , RNA Nucleotidyltransferases/antagonists & inhibitors , RNA Nucleotidyltransferases/genetics , RNA, Viral/genetics
6.
Mol Ther ; 29(10): 2910-2919, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34091052

ABSTRACT

N-Acetylgalactosamine (GalNAc) conjugated short interfering RNAs (siRNAs) are a leading RNA interference (RNAi) platform allowing targeted inhibition of disease-causing genes in hepatocytes. More than a decade of development has recently resulted in the first approvals for this class of drugs. While substantial effort has been made to improve nucleic acid modification patterns for better payload stability and efficacy, relatively little attention has been given to the GalNAc targeting ligand. In addition, the lack of an intrinsic endosomal release mechanism has limited potency. Here, we report a stepwise analysis of the structure activity relationships (SAR) of the components comprising these targeting ligands. We show that there is relatively little difference in biological performance between bi-, tri-, and tetravalent ligand structures while identifying other features that affect their biological activity more significantly. Further, we demonstrate that subcutaneous co-administration of a GalNAc-functionalized, pH responsive endosomal release agent markedly improved the activity and duration of effect for siRNA conjugates, without compromising tolerability, in non-human primates. These findings could address a significant bottleneck for future siRNA ligand conjugate development.


Subject(s)
Acetylgalactosamine/chemistry , Asialoglycoprotein Receptor/metabolism , RNA, Small Interfering/administration & dosage , Animals , Female , Hep G2 Cells , Humans , Injections, Subcutaneous , Ligands , Liposomes , Male , Mice , Nanoparticles , Primates , RNA, Small Interfering/chemistry , Structure-Activity Relationship
7.
Nat Commun ; 12(1): 1222, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619272

ABSTRACT

Programmed death-ligand 1 is a glycoprotein expressed on antigen presenting cells, hepatocytes, and tumors which upon interaction with programmed death-1, results in inhibition of antigen-specific T cell responses. Here, we report a mechanism of inhibiting programmed death-ligand 1 through small molecule-induced dimerization and internalization. This represents a mechanism of checkpoint inhibition, which differentiates from anti-programmed death-ligand 1 antibodies which function through molecular disruption of the programmed death 1 interaction. Testing of programmed death ligand 1 small molecule inhibition in a humanized mouse model of colorectal cancer results in a significant reduction in tumor size and promotes T cell proliferation. In addition, antigen-specific T and B cell responses from patients with chronic hepatitis B infection are significantly elevated upon programmed death ligand 1 small molecule inhibitor treatment. Taken together, these data identify a mechanism of small molecule-induced programmed death ligand 1 internalization with potential therapeutic implications in oncology and chronic viral infections.


Subject(s)
B7-H1 Antigen/metabolism , Endocytosis , Immune Checkpoint Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , CHO Cells , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Cricetulus , Disease Models, Animal , Female , Hepatitis B virus/drug effects , Humans , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , Protein Multimerization/drug effects , Small Molecule Libraries/chemistry
8.
Protein Expr Purif ; 179: 105796, 2021 03.
Article in English | MEDLINE | ID: mdl-33221505

ABSTRACT

TREM2 has been identified by genomic analysis as a potential and novel target for the treatment of Alzheimer's disease. To enable structure-based screening of potential small molecule therapeutics, we sought to develop a robust crystallization platform for the TREM2 Ig-like domain. A systematic set of constructs containing the structural chaperone, maltose binding protein (MBP), fused to the Ig domain of TREM2, were evaluated in parallel expression and purification, followed by crystallization studies. Using protein crystallization and high-resolution diffraction as a readout, a MBP-TREM2 Ig fusion construct was identified that generates reproducible protein crystals diffracting at 2.0 Å, which makes it suitable for soaking of potential ligands. Importantly, analysis of crystal packing interfaces indicates that most of the surface of the TREM2 Ig domain is available for small molecule binding. A proof of concept co-crystallization study with a small library of fragments validated potential utility of this system for the discovery of new TREM2 therapeutics.


Subject(s)
Crystallization/methods , Membrane Glycoproteins , Molecular Chaperones , Receptors, Immunologic , Recombinant Fusion Proteins , Humans , Maltose-Binding Proteins/chemistry , Maltose-Binding Proteins/genetics , Maltose-Binding Proteins/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
10.
ACS Infect Dis ; 5(5): 738-749, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30408957

ABSTRACT

Hepatitis delta virus (HDV) infects 10-20 million individuals worldwide and causes severe fulminant hepatitis with high likelihood of cirrhosis and hepatocellular carcinoma. HDV infection cannot occur in the absence of the surface antigen (HBsAg) of the hepatitis B virus. RNA interference is an effective mechanism by which to inhibit viral transcripts, and siRNA therapeutics sharing this mechanism have begun to demonstrate clinical efficacy. Here we assessed the outcome of HBV-targeting siRNA intervention against HDV and compared it to a direct anti-HDV siRNA approach in dually infected humanized mice. Treatment with ARB-1740, a clinical stage HBV-targeting siRNA agent delivered using lipid nanoparticle (LNP) technology, effectively reduced HBV viremia by 2.3 log10 and serum HBsAg by 2.6 log10, leading to 1.6 log10 reduction of HDV viremia. In contrast, HDV-targeting siRNA inhibited HDV in both blood and liver compartments without affecting HBV and PEGylated interferon-alpha reduced HBV viremia by 2.0 log10 but had no effect on HDV viremia under these study conditions. These results illustrate the inhibitory effects of siRNAs against these two viral infections and suggest that ARB-1740 may be of therapeutic benefit for hepatitis delta patients, a subpopulation with high unmet medical need.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis D/drug therapy , Hepatitis Delta Virus/drug effects , RNA Interference , Animals , Animals, Genetically Modified , Disease Models, Animal , Female , Hepatitis B virus/drug effects , Humans , Mice
11.
ACS Infect Dis ; 5(5): 725-737, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30403127

ABSTRACT

Current approved nucleoside analogue treatments for chronic hepatitis B virus (HBV) infection are effective at controlling viral titer but are not curative and have minimal impact on the production of viral proteins such as surface antigen (HBsAg), the HBV envelope protein believed to play a role in maintaining the immune tolerant state required for viral persistence. Novel agents are needed to effect HBV cure, and reduction of HBV antigenemia may potentiate activation of effective and long-lasting host immune control. ARB-1740 is a clinical stage RNA interference agent composed of three siRNAs delivered using lipid nanoparticle technology. In a number of cell and animal models of HBV, ARB-1740 caused HBV RNA reduction, leading to inhibition of multiple elements of the viral life cycle including HBsAg, HBeAg, and HBcAg viral proteins as well as replication marker HBV DNA. ARB-1740 demonstrated pan-genotypic activity in vitro and in vivo, targeting three distinct highly conserved regions of the HBV genome, and effectively inhibited replication of nucleoside analogue-resistant HBV variants. Combination of ARB-1740 with a capsid inhibitor and pegylated interferon-alpha led to greater liver HBsAg reduction which correlated with more robust induction of innate immune responses in a human chimeric mouse model of HBV. The preclinical profile of ARB-1740 demonstrates the promise of RNA interference and HBV antigen reduction in treatment strategies driving toward a cure for HBV.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis B virus/drug effects , Hepatitis B, Chronic/drug therapy , RNA Interference , RNA, Small Interfering/therapeutic use , Animals , Genome, Viral , Humans , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/chemistry , RNA, Small Interfering/chemistry , Virus Replication/drug effects
12.
Article in English | MEDLINE | ID: mdl-29555628

ABSTRACT

AB-423 is a member of the sulfamoylbenzamide (SBA) class of hepatitis B virus (HBV) capsid inhibitors in phase 1 clinical trials. In cell culture models, AB-423 showed potent inhibition of HBV replication (50% effective concentration [EC50] = 0.08 to 0.27 µM; EC90 = 0.33 to 1.32 µM) with no significant cytotoxicity (50% cytotoxic concentration > 10 µM). Addition of 40% human serum resulted in a 5-fold increase in the EC50s. AB-423 inhibited HBV genotypes A through D and nucleos(t)ide-resistant variants in vitro Treatment of HepDES19 cells with AB-423 resulted in capsid particles devoid of encapsidated pregenomic RNA and relaxed circular DNA (rcDNA), indicating that it is a class II capsid inhibitor. In a de novo infection model, AB-423 prevented the conversion of encapsidated rcDNA to covalently closed circular DNA, presumably by interfering with the capsid uncoating process. Molecular docking of AB-423 into crystal structures of heteroaryldihydropyrimidines and an SBA and biochemical studies suggest that AB-423 likely also binds to the dimer-dimer interface of core protein. In vitro dual combination studies with AB-423 and anti-HBV agents, such as nucleos(t)ide analogs, RNA interference agents, or interferon alpha, resulted in additive to synergistic antiviral activity. Pharmacokinetic studies with AB-423 in CD-1 mice showed significant systemic exposures and higher levels of accumulation in the liver. A 7-day twice-daily administration of AB-423 in a hydrodynamic injection mouse model of HBV infection resulted in a dose-dependent reduction in serum HBV DNA levels, and combination with entecavir or ARB-1467 resulted in a trend toward antiviral activity greater than that of either agent alone, consistent with the results of the in vitro combination studies. The overall preclinical profile of AB-423 supports its further evaluation for safety, pharmacokinetics, and antiviral activity in patients with chronic hepatitis B.


Subject(s)
Antiviral Agents/pharmacology , Capsid/metabolism , Hepatitis B virus/drug effects , Hepatitis B/drug therapy , Virus Assembly/drug effects , Animals , Binding Sites , Cell Line, Tumor , DNA, Circular/metabolism , DNA, Viral/blood , DNA, Viral/metabolism , Female , Guanine/analogs & derivatives , Guanine/pharmacology , Hepatitis B virus/growth & development , Humans , Mice , Molecular Docking Simulation , Protein Binding , RNA, Viral/genetics
13.
Antiviral Res ; 149: 191-201, 2018 01.
Article in English | MEDLINE | ID: mdl-29133129

ABSTRACT

In pursuit of novel therapeutics targeting the hepatitis B virus (HBV) infection, we evaluated a dihydroquinolizinone compound (DHQ-1) that in the nanomolar range reduced the production of virion and surface protein (HBsAg) in tissue culture. This compound also showed broad HBV genotype coverage, but was inactive against a panel of DNA and RNA viruses of other species. Oral administration of DHQ-1 in the AAV-HBV mouse model resulted in a significant reduction of serum HBsAg as soon as 4 days following the commencement of treatment. Reduction of HBV markers in both in vitro and in vivo experiments was related to the reduced amount of viral RNA including pre-genomic RNA (pgRNA) and 2.4/2.1 kb HBsAg mRNA. Nuclear run-on and subcellular fractionation experiments indicated that DHQ-1 mediated HBV RNA reduction was the result of accelerated viral RNA degradation in the nucleus, rather than the consequence of inhibition of transcription initiation. Through mutagenesis of HBsAg gene sequences, we found induction of HBsAg mRNA decay by DHQ-1 required the presence of the HBV posttranscriptional regulatory element (HPRE), with a 109 nucleotides sequence within the central region of the HPRE alpha sub-element being the most critical. Taken together, the current study shows that a small molecule can reduce the overall levels of HBV RNA, especially the HBsAg mRNA, and viral surface proteins. This may shed light on the development of a new class of HBV therapeutics.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B Surface Antigens/genetics , Hepatitis B virus/drug effects , Hepatitis B virus/genetics , RNA Processing, Post-Transcriptional/drug effects , RNA, Messenger/genetics , RNA, Viral/genetics , Response Elements , Binding Sites , Genotype , Humans , Protein Binding , RNA Stability/drug effects , Transfection , Virus Replication
14.
Nat Microbiol ; 1(10): 16142, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27670117

ABSTRACT

Although significant progress has been made in developing therapeutics against Zaire ebolavirus, these therapies do not protect against other Ebola species such as Sudan ebolavirus (SUDV). Here, we describe an RNA interference therapeutic comprising siRNA targeting the SUDV VP35 gene encapsulated in lipid nanoparticle (LNP) technology with increased potency beyond formulations used in TKM-Ebola clinical trials. Twenty-five rhesus monkeys were challenged with a lethal dose of SUDV. Twenty animals received siRNA-LNP beginning at 1, 2, 3, 4 or 5 days post-challenge. VP35-targeting siRNA-LNP treatment resulted in up to 100% survival, even when initiated when fever, viraemia and disease signs were evident. Treatment effectively controlled viral replication, mediating up to 4 log10 reductions after dosing. Mirroring clinical findings, a correlation between high viral loads and fatal outcome was observed, emphasizing the importance of stratifying efficacy according to viral load. In summary, strong survival benefit and rapid control of SUDV replication by VP35-targeting LNP confirm its therapeutic potential in combatting this lethal disease.


Subject(s)
Hemorrhagic Fever, Ebola/therapy , Lipids , RNA Interference , RNA, Small Interfering/therapeutic use , Animals , Antibodies, Viral , Disease Models, Animal , Drug Compounding , Ebolavirus/isolation & purification , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/virology , Hep G2 Cells , Humans , Macaca mulatta , Nanoparticles/administration & dosage , Nanoparticles/chemistry , RNA, Small Interfering/genetics , Sudan , Viral Load/drug effects , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Viremia/therapy , Virus Replication
15.
J Infect Dis ; 214(suppl 3): S367-S374, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27571900

ABSTRACT

BACKGROUND: Convalescent serum and blood were used to treat patients during outbreaks of Zaire ebolavirus (ZEBOV) infection in 1976 and 1995, with inconclusive results. During the recent 2013-2016 West African epidemic, serum/plasma from survivors of ZEBOV infection was used to treat patients in the affected countries and several repatriated patients. The effectiveness of this strategy remains unknown. METHODS: Nine rhesus monkeys were experimentally infected with ZEBOV-Makona. Beginning on day 3 after exposure (at the onset of viremia), 4 animals were treated with homologous ZEBOV-Makona convalescent macaque sera, 3 animals were treated in parallel with heterologous Sudan ebolavirus (SEBOV) convalescent macaque sera, and 2 animals served as positive controls and were not treated. Surviving animals received additional treatments on days 6 and 9. RESULTS: Both untreated control animals died on postinfection day 9. All 4 ZEBOV-Makona-infected macaques treated with homologous ZEBOV-Makona convalescent sera died on days 8-9. One macaque treated with heterologous SEBOV convalescent sera survived, while the other animals treated with the heterologous SEBOV sera died on days 7 and 9. CONCLUSIONS: The findings suggest that convalescent sera alone is not sufficient for providing 100% protection against lethal ZEBOV infection when administered at the onset of viremia.


Subject(s)
Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Immunization, Passive , Animals , Convalescence , Female , Hemorrhagic Fever, Ebola/virology , Humans , Macaca mulatta , Male , Serum/immunology , Viremia
16.
Nature ; 521(7552): 362-5, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25901685

ABSTRACT

The current outbreak of Ebola virus in West Africa is unprecedented, causing more cases and fatalities than all previous outbreaks combined, and has yet to be controlled. Several post-exposure interventions have been employed under compassionate use to treat patients repatriated to Europe and the United States. However, the in vivo efficacy of these interventions against the new outbreak strain of Ebola virus is unknown. Here we show that lipid-nanoparticle-encapsulated short interfering RNAs (siRNAs) rapidly adapted to target the Makona outbreak strain of Ebola virus are able to protect 100% of rhesus monkeys against lethal challenge when treatment was initiated at 3 days after exposure while animals were viraemic and clinically ill. Although all infected animals showed evidence of advanced disease including abnormal haematology, blood chemistry and coagulopathy, siRNA-treated animals had milder clinical features and fully recovered, while the untreated control animals succumbed to the disease. These results represent the first, to our knowledge, successful demonstration of therapeutic anti-Ebola virus efficacy against the new outbreak strain in nonhuman primates and highlight the rapid development of lipid-nanoparticle-delivered siRNA as a countermeasure against this highly lethal human disease.


Subject(s)
Ebolavirus/drug effects , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/therapy , Hemorrhagic Fever, Ebola/virology , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , Animals , Base Sequence , Disease Models, Animal , Ebolavirus/classification , Female , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/prevention & control , Humans , Macaca mulatta/virology , Male , RNA, Small Interfering/pharmacology , Survival Analysis , Time Factors , Treatment Outcome , Viral Load/drug effects
17.
Ann Glob Health ; 81(5): 718-24, 2015.
Article in English | MEDLINE | ID: mdl-27036731

ABSTRACT

BACKGROUND: Worldwide, there is a gap between the burden of mental distress and disorder and access to mental health care. This gap is particularly large in low- and middle-income countries (LMICs). After the 2010 earthquake in Haiti, the international health care organizations Partners in Health and Zanmi Lasante worked to expand local mental health services in rural Haiti. OBJECTIVE: The aims of this study are to describe clinical characteristics of the patients served during a pilot project to deliver community-based psychiatric services in rural Haiti and to show how this experience complements the Mental Health Gap Action Programme ("mhGAP"), a tool developed by the World Health Organization to support mental health care delivery by nonspecialists in LMICs. METHODS: The pilot was conducted in March 2011. A visiting psychiatrist traveled to rural Haiti and paired with local clinicians to evaluate patients and to support quality improvement practices in psychiatric care. Patients received a standard neuropsychiatric evaluation. mhGAP was an important clinical reference. To assess the experience, we conducted a retrospective chart review of outpatient encounters. FINDINGS: Sixty-five patients presented with a wide range of common psychiatric, neurologic, and general medical conditions. Forty-nine of these patients (75%) reported primary problems subsumed by an mhGAP module. Fifteen patients (23%) reported headache as their chief complain, a condition that is not currently covered by mhGAP. Surprisingly, only 3 patients (5%), reported earthquake-related distress. CONCLUSIONS: Our clinical data reinforce the need for provision of standard psychiatric and neurologic services in LMICs. Such services ought to accompany interventions targeted specifically at disaster-related problems. Clinical situations falling outside existing mhGAP modules inspired the development of supplemental treatment protocols. These observations informed coordinated efforts at Zanmi Lasante to build a sustainable, integrated mental health system in Haiti that may be relevant to other resource-limited settings.


Subject(s)
Community Mental Health Services/methods , Delivery of Health Care/methods , Disasters , Earthquakes , Mental Disorders/therapy , Rural Population , Bipolar Disorder/therapy , Dementia/therapy , Depression/therapy , Developing Countries , Haiti , Headache/therapy , Humans , Neurology , Pilot Projects , Psychiatry , Psychotic Disorders/therapy , Quality Improvement , Retrospective Studies , Seizures/therapy , Stress, Psychological/therapy
18.
Sci Transl Med ; 6(250): 250ra116, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25143366

ABSTRACT

Marburg virus (MARV) and the closely related filovirus Ebola virus cause severe and often fatal hemorrhagic fever (HF) in humans and nonhuman primates with mortality rates up to 90%. There are no vaccines or drugs approved for human use, and no postexposure treatment has completely protected nonhuman primates against MARV-Angola, the strain associated with the highest rate of mortality in naturally occurring human outbreaks. Studies performed with other MARV strains assessed candidate treatments at times shortly after virus exposure, before signs of disease are detectable. We assessed the efficacy of lipid nanoparticle (LNP) delivery of anti-MARV nucleoprotein (NP)-targeting small interfering RNA (siRNA) at several time points after virus exposure, including after the onset of detectable disease in a uniformly lethal nonhuman primate model of MARV-Angola HF. Twenty-one rhesus monkeys were challenged with a lethal dose of MARV-Angola. Sixteen of these animals were treated with LNP containing anti-MARV NP siRNA beginning at 30 to 45 min, 1 day, 2 days, or 3 days after virus challenge. All 16 macaques that received LNP-encapsulated anti-MARV NP siRNA survived infection, whereas the untreated or mock-treated control subjects succumbed to disease between days 7 and 9 after infection. These results represent the successful demonstration of therapeutic anti-MARV-Angola efficacy in nonhuman primates and highlight the substantial impact of an LNP-delivered siRNA therapeutic as a countermeasure against this highly lethal human disease.


Subject(s)
Lipids/therapeutic use , Macaca mulatta/virology , Marburg Virus Disease/virology , Marburgvirus/physiology , Nanoparticles/therapeutic use , RNA, Small Interfering/therapeutic use , Animals , Antigens, Viral/immunology , Humans , Macaca mulatta/immunology , Marburg Virus Disease/pathology , Marburg Virus Disease/therapy , Marburgvirus/immunology , Nanoparticles/chemistry , RNA, Viral/metabolism , Survival Analysis , Treatment Outcome , Viremia/pathology
20.
Neurology ; 80(22): 2062-4, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23713085

ABSTRACT

The idea of a Global Health Section within the American Academy of Neurology (AAN) came from a group of neurologists with active work in sub-Saharan Africa, who believed that the AAN could provide a greater leadership role in supporting the advancement of quality neurologic training, research, and patient care in low and middle-income countries (LMICs). Initially a Special Interest Group, the Global Health Section was approved for full section status in September 2011 and endorsed by the AAN Board of Directors in October 2011. The Global Health Section currently consists of over 200 members. In a 2-part series, we present a summary of the Global Health Section strategic plan and vision for future activities.


Subject(s)
Developing Countries , Global Health , Neurology/organization & administration , Societies, Medical/organization & administration , Developing Countries/economics , Developing Countries/statistics & numerical data , Global Health/economics , Global Health/education , Global Health/standards , Global Health/statistics & numerical data , Humans , Nervous System Diseases/economics , Nervous System Diseases/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...