Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Krystle N. Agans is active.

Publication


Featured researches published by Krystle N. Agans.


Nature | 2015

Lipid nanoparticle siRNA treatment of Ebola virus Makona infected nonhuman primates

Emily P. Thi; Chad E. Mire; Amy Lee; Joan B. Geisbert; Joy Z. Zhou; Krystle N. Agans; Nicholas M. Snead; Daniel J. Deer; Trisha R. Barnard; Karla A. Fenton; Ian Maclachlan; Thomas W. Geisbert

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.


Nature | 2015

Single-dose attenuated Vesiculovax vaccines protect primates against Ebola Makona virus

Chad E. Mire; Demetrius Matassov; Joan B. Geisbert; Theresa Latham; Krystle N. Agans; Rong Xu; Ayuko Ota-Setlik; Michael A. Egan; Karla A. Fenton; David K. Clarke; John H. Eldridge; Thomas W. Geisbert

The family Filoviridae contains three genera, Ebolavirus (EBOV), Marburg virus, and Cuevavirus. Some members of the EBOV genus, including Zaire ebolavirus (ZEBOV), can cause lethal haemorrhagic fever in humans. During 2014 an unprecedented ZEBOV outbreak occurred in West Africa and is still ongoing, resulting in over 10,000 deaths, and causing global concern of uncontrolled disease. To meet this challenge a rapid-acting vaccine is needed. Many vaccine approaches have shown promise in being able to protect nonhuman primates against ZEBOV. In response to the current ZEBOV outbreak several of these vaccines have been fast tracked for human use. However, it is not known whether any of these vaccines can provide protection against the new outbreak Makona strain of ZEBOV. One of these approaches is a first-generation recombinant vesicular stomatitis virus (rVSV)-based vaccine expressing the ZEBOV glycoprotein (GP) (rVSV/ZEBOV). To address safety concerns associated with this vector, we developed two candidate, further-attenuated rVSV/ZEBOV vaccines. Both attenuated vaccines produced an approximately tenfold lower vaccine-associated viraemia compared to the first-generation vaccine and both provided complete, single-dose protection of macaques from lethal challenge with the Makona outbreak strain of ZEBOV.


Science Translational Medicine | 2014

Marburg virus infection in nonhuman primates: Therapeutic treatment by lipid-encapsulated siRNA.

Emily P. Thi; Chad E. Mire; Raul Ursic-Bedoya; Joan B. Geisbert; Amy C. H. Lee; Krystle N. Agans; Marjorie Robbins; Daniel J. Deer; Karla A. Fenton; Ian Maclachlan; Thomas W. Geisbert

Lipid nanoparticle delivery of anti-MARV nucleoprotein–targeting small interfering RNA can treat Marburg virus in nonhuman primates after symptom onset. Medicating Marburg Marburg virus (MARV) and the closely related filovirus Ebola virus cause severe and often fatal hemorrhagic fever; however, there are currently no vaccines or drugs approved for human use to treat these devastating infections. Thi et al. now report that a lipid-encapsulated siRNA can treat MARV in nonhuman primates after symptom onset. They use lipid nanoparticles to deliver siRNA targeting the MARV nucleoprotein to treated animals at various time points after virus exposure. All animals that received the therapy survived MARV infection, including those that were treated 3 days after infection—a stage when animals are viremic and demonstrate the first clinical signs of disease. Therefore, this approach holds promise as a strategy to treat filovirus infection in humans. 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.


Science Translational Medicine | 2014

Therapeutic treatment of Nipah virus infection in nonhuman primates with a neutralizing human monoclonal antibody.

Thomas W. Geisbert; Chad E. Mire; Joan B. Geisbert; Yee Peng Chan; Krystle N. Agans; Friederike Feldmann; Karla A. Fenton; Zhongyu Zhu; Dimiter S. Dimitrov; Dana P. Scott; Katharine N. Bossart; Heinz Feldmann; Christopher C. Broder

A monoclonal antibody against Nipah virus has in vivo efficacy in a nonhuman primate. Time’s Up for Nipah Virus A human antibody may be the key for treating a deadly viral infection. Geisbert et al. report that a human monoclonal antibody to the emerging zoonotic paramyxovirus Nipah virus (NiV) can treat nonhuman primates at the onset of clinical symptoms. Fruit bats are the natural reservoir for NiV. However, NiV can infect a broad range of mammalian species, and exposed pigs may serve as an intermediary for human infection, which is frequently fatal. There is currently no approved human treatment for NiV, and previous studies have only shown efficacy after infection—but before the onset of clinical symptoms. Now, a human monoclonal antibody has resulted in survival in all animals treated, even after the development of clinical symptoms. This antibody represents a potential new therapeutic strategy for this emerging viral infection. Nipah virus (NiV) is an emerging zoonotic paramyxovirus that causes severe and often fatal disease in pigs and humans. There are currently no vaccines or treatments approved for human use. Studies in small-animal models of NiV infection suggest that antibody therapy may be a promising treatment. However, most studies have assessed treatment at times shortly after virus exposure before animals show signs of disease. We assessed the efficacy of a fully human monoclonal antibody, m102.4, at several time points after virus exposure including at the onset of clinical illness in a uniformly lethal nonhuman primate model of NiV disease. Sixteen African green monkeys (AGMs) were challenged intratracheally with a lethal dose of NiV, and 12 animals were infused twice with m102.4 (15 mg/kg) beginning at either 1, 3, or 5 days after virus challenge and again about 2 days later. The presence of viral RNA, infectious virus, and/or NiV-specific immune responses demonstrated that all subjects were infected after challenge. All 12 AGMs that received m102.4 survived infection, whereas the untreated control subjects succumbed to disease between days 8 and 10 after infection. AGMs in the day 5 treatment group exhibited clinical signs of disease, but all animals recovered by day 16. These results represent the successful therapeutic in vivo efficacy by an investigational drug against NiV in a nonhuman primate and highlight the potential impact that a monoclonal antibody can have on a highly pathogenic zoonotic human disease.


PLOS ONE | 2014

Durability of a vesicular stomatitis virus-based marburg virus vaccine in nonhuman primates.

Chad E. Mire; Joan B. Geisbert; Krystle N. Agans; Benjamin A. Satterfield; Krista Versteeg; Elizabeth A. Fritz; Heinz Feldmann; Lisa Hensley; Thomas W. Geisbert

The filoviruses, Marburg virus (MARV) and Ebola virus, causes severe hemorrhagic fever with high mortality in humans and nonhuman primates. A promising filovirus vaccine under development is based on a recombinant vesicular stomatitis virus (rVSV) that expresses individual filovirus glycoproteins (GPs) in place of the VSV glycoprotein (G). These vaccines have shown 100% efficacy against filovirus infection in nonhuman primates when challenge occurs 28–35 days after a single injection immunization. Here, we examined the ability of a rVSV MARV-GP vaccine to provide protection when challenge occurs more than a year after vaccination. Cynomolgus macaques were immunized with rVSV-MARV-GP and challenged with MARV approximately 14 months after vaccination. Immunization resulted in the vaccine cohort of six animals having anti-MARV GP IgG throughout the pre-challenge period. Following MARV challenge none of the vaccinated animals showed any signs of clinical disease or viremia and all were completely protected from MARV infection. Two unvaccinated control animals exhibited signs consistent with MARV infection and both succumbed. Importantly, these data are the first to show 100% protective efficacy against any high dose filovirus challenge beyond 8 weeks after final vaccination. These findings demonstrate the durability of VSV-based filovirus vaccines.


The Journal of Infectious Diseases | 2015

A Single-Vector, Single-Injection Trivalent Filovirus Vaccine: Proof of Concept Study in Outbred Guinea Pigs

Chad E. Mire; Joan B. Geisbert; Krista Versteeg; Natalia Mamaeva; Krystle N. Agans; Thomas W. Geisbert; John H. Connor

The filoviruses, Marburg marburgvirus (MARV), Zaire ebolavirus (ZEBOV), and Sudan ebolavirus (SEBOV), cause severe and often fatal hemorrhagic fever in humans and nonhuman primates (NHPs). Monovalent recombinant vesicular stomatitis virus (rVSV)-based vaccine vectors, which encode a filovirus glycoprotein (GP) in place of the VSV glycoprotein, have shown 100% efficacy against homologous filovirus challenge in rodent and NHP studies. Here, we examined the utility of a single-vector, single-injection trivalent rVSV vector expressing MARV, ZEBOV, and SEBOV GPs to protect against MARV-, ZEBOV-, and SEBOV-induced disease in outbred Hartley guinea pigs where we observed protection from effects of all 3 filoviruses.


Mbio | 2016

Polyamines and Hypusination Are Required for Ebolavirus Gene Expression and Replication

Michelle E. Olsen; Claire Marie Filone; Dan Rozelle; Chad E. Mire; Krystle N. Agans; Lisa E. Hensley; John H. Connor

ABSTRACT Ebolavirus (EBOV) is an RNA virus that is known to cause severe hemorrhagic fever in humans and other primates. EBOV successfully enters and replicates in many cell types. This replication is dependent on the virus successfully coopting a number of cellular factors. Many of these factors are currently unidentified but represent potential targets for antiviral therapeutics. Here we show that cellular polyamines are critical for EBOV replication. We found that small-molecule inhibitors of polyamine synthesis block gene expression driven by the viral RNA-dependent RNA polymerase. Short hairpin RNA (shRNA) knockdown of the polyamine pathway enzyme spermidine synthase also resulted in reduced EBOV replication. These findings led us to further investigate spermidine, a polyamine that is essential for the hypusination of eukaryotic initiation factor 5A (eIF5A). Blocking the hypusination of eIF5A (and thereby inhibiting its function) inhibited both EBOV gene expression and viral replication. The mechanism appears to be due to the importance of hypusinated eIF5A for the accumulation of VP30, an essential component of the viral polymerase. The same reduction in hypusinated eIF5A did not alter the accumulation of other viral polymerase components. This action makes eIF5A function an important gate for proper EBOV polymerase assembly and function through the control of a single virus protein. IMPORTANCE Ebolavirus (EBOV) is one of the most lethal human pathogens known. EBOV requires host factors for replication due to its small RNA genome. Here we show that the host protein eIF5A in its activated form is necessary for EBOV replication. We further show that the mechanism is through the accumulation of a single EBOV protein, VP30. To date, no other host proteins have been shown to interfere with the translation or stability of an EBOV protein. Activated eIF5A is the only protein in the cell known to contain the specific modification of hypusine; therefore, this pathway is a target for drug development. Further investigation into the mechanism of eIF5A interaction with VP30 could provide insight into therapeutics to combat EBOV. Ebolavirus (EBOV) is one of the most lethal human pathogens known. EBOV requires host factors for replication due to its small RNA genome. Here we show that the host protein eIF5A in its activated form is necessary for EBOV replication. We further show that the mechanism is through the accumulation of a single EBOV protein, VP30. To date, no other host proteins have been shown to interfere with the translation or stability of an EBOV protein. Activated eIF5A is the only protein in the cell known to contain the specific modification of hypusine; therefore, this pathway is a target for drug development. Further investigation into the mechanism of eIF5A interaction with VP30 could provide insight into therapeutics to combat EBOV.


The Journal of Infectious Diseases | 2016

Passive Immunotherapy: Assessment of Convalescent Serum Against Ebola Virus Makona Infection in Nonhuman Primates

Chad E. Mire; Joan B. Geisbert; Krystle N. Agans; Emily P. Thi; Amy Lee; Karla A. Fenton; Thomas W. Geisbert

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.


Nature Communications | 2015

The immunomodulating V and W proteins of Nipah virus determine disease course

Benjamin A. Satterfield; Robert W. Cross; Karla A. Fenton; Krystle N. Agans; Christopher F. Basler; Thomas W. Geisbert; Chad E. Mire

The viral determinants that contribute to Nipah virus (NiV)-mediated disease are poorly understood compared with other paramyxoviruses. Here we use recombinant NiVs (rNiVs) to examine the contributions of the NiV V and W proteins to NiV pathogenesis in a ferret model. We show that a V-deficient rNiV is susceptible to the innate immune response in vitro and behaves as a replicating non-lethal virus in vivo. Remarkably, rNiV lacking W expression results in a delayed and altered disease course with decreased respiratory disease and increased terminal neurological disease associated with altered in vitro inflammatory cytokine production. This study confirms the V protein as the major determinant of pathogenesis, also being the first in vivo study to show that the W protein modulates the inflammatory host immune response in a manner that determines the disease course.


Scientific Reports | 2016

Pathogenic Differences between Nipah Virus Bangladesh and Malaysia Strains in Primates: Implications for Antibody Therapy

Chad E. Mire; Benjamin A. Satterfield; Joan B. Geisbert; Krystle N. Agans; Viktoriya Borisevich; Lianying Yan; Yee Peng Chan; Robert W. Cross; Karla A. Fenton; Christopher C. Broder; Thomas W. Geisbert

Nipah virus (NiV) is a paramyxovirus that causes severe disease in humans and animals. There are two distinct strains of NiV, Malaysia (NiVM) and Bangladesh (NiVB). Differences in transmission patterns and mortality rates suggest that NiVB may be more pathogenic than NiVM. To investigate pathogenic differences between strains, 4 African green monkeys (AGM) were exposed to NiVM and 4 AGMs were exposed to NiVB. While NiVB was uniformly lethal, only 50% of NiVM-infected animals succumbed to infection. Histopathology of lungs and spleens from NiVB-infected AGMs was significantly more severe than NiVM-infected animals. Importantly, a second study utilizing 11 AGMs showed that the therapeutic window for human monoclonal antibody m102.4, previously shown to rescue AGMs from NiVM infection, was much shorter in NiVB-infected AGMs. Together, these data show that NiVB is more pathogenic in AGMs under identical experimental conditions and suggests that postexposure treatments may need to be NiV strain specific for optimal efficacy.

Collaboration


Dive into the Krystle N. Agans's collaboration.

Top Co-Authors

Avatar

Chad E. Mire

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Thomas W. Geisbert

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Joan B. Geisbert

United States Department of the Army

View shared research outputs
Top Co-Authors

Avatar

Karla A. Fenton

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Robert W. Cross

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Daniel J. Deer

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Viktoriya Borisevich

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin A. Satterfield

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge