Network


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

Hotspot


Dive into the research topics where Karla A. Fenton is active.

Publication


Featured researches published by Karla A. Fenton.


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.


Journal of Clinical Investigation | 2015

Aerosolized Ebola vaccine protects primates and elicits lung-resident T cell responses

Michelle Meyer; Tania Garron; Ndongala Michel Lubaki; Chad E. Mire; Karla A. Fenton; Curtis Klages; Gene G. Olinger; Thomas W. Geisbert; Peter L. Collins; Alexander Bukreyev

Direct delivery of aerosolized vaccines to the respiratory mucosa elicits both systemic and mucosal responses. This vaccine strategy has not been tested for Ebola virus (EBOV) or other hemorrhagic fever viruses. Here, we examined the immunogenicity and protective efficacy of an aerosolized human parainfluenza virus type 3-vectored vaccine that expresses the glycoprotein (GP) of EBOV (HPIV3/EboGP) delivered to the respiratory tract. Rhesus macaques were vaccinated with aerosolized HPIV3/EboGP, liquid HPIV3/EboGP, or an unrelated, intramuscular, Venezuelan equine encephalitis replicon vaccine expressing EBOV GP. Serum and mucosal samples from aerosolized HPIV3/EboGP recipients exhibited high EBOV-specific IgG, IgA, and neutralizing antibody titers, which exceeded or equaled titers observed in liquid recipients. The HPIV3/EboGP vaccine induced an EBOV-specific cellular response that was greatest in the lungs and yielded polyfunctional CD8+ T cells, including a subset that expressed CD103 (αE integrin), and CD4+ T helper cells that were predominately type 1. The magnitude of the CD4+ T cell response was greater in aerosol vaccinees. The HPIV3/EboGP vaccine produced a more robust cell-mediated and humoral immune response than the systemic replicon vaccine. Moreover, 1 aerosol HPIV3/EboGP dose conferred 100% protection to macaques exposed to EBOV. Aerosol vaccination represents a useful and feasible vaccination mode that can be implemented with ease in a filovirus disease outbreak situation.


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.


Nature Medicine | 2017

A chikungunya fever vaccine utilizing an insect-specific virus platform

Jesse H. Erasmus; Albert J. Auguste; Jason T. Kaelber; Huanle Luo; Shannan L. Rossi; Karla A. Fenton; Grace Leal; Dal Young Kim; Wah Chiu; Tian Wang; Ilya Frolov; Farooq Nasar; Scott C. Weaver

Traditionally, vaccine development involves tradeoffs between immunogenicity and safety. Live-attenuated vaccines typically offer rapid and durable immunity but have reduced safety when compared to inactivated vaccines. In contrast, the inability of inactivated vaccines to replicate enhances safety at the expense of immunogenicity, often necessitating multiple doses and boosters. To overcome these tradeoffs, we developed the insect-specific alphavirus, Eilat virus (EILV), as a vaccine platform. To address the chikungunya fever (CHIKF) pandemic, we used an EILV cDNA clone to design a chimeric virus containing the chikungunya virus (CHIKV) structural proteins. The recombinant EILV/CHIKV was structurally identical at 10 Å to wild-type CHIKV, as determined by single-particle cryo-electron microscopy, and it mimicked the early stages of CHIKV replication in vertebrate cells from attachment and entry to viral RNA delivery. Yet the recombinant virus remained completely defective for productive replication, providing a high degree of safety. A single dose of EILV/CHIKV produced in mosquito cells elicited rapid (within 4 d) and long-lasting (>290 d) neutralizing antibodies that provided complete protection in two different mouse models. In nonhuman primates, EILV/CHIKV elicited rapid and robust immunity that protected against viremia and telemetrically monitored fever. Our EILV platform represents the first structurally native application of an insect-specific virus in preclinical vaccine development and highlights the potential application of such viruses in vaccinology.


The Journal of Infectious Diseases | 2016

The Domestic Ferret (Mustela putorius furo) as a Lethal Infection Model for 3 Species of Ebolavirus

Robert W. Cross; Chad E. Mire; Viktoriya Borisevich; Joan B. Geisbert; Karla A. Fenton; Thomas W. Geisbert

Small-animal models have been developed for several Filoviridae species; however, serial adaptation was required to produce lethal infection. These adapted viruses have sequence changes in several genes, including those that modulate the host immune response. Nonhuman primate models do not require adaptation of filoviruses. Here, we describe lethal models of disease for Bundibugyo, Sudan, and Zaire species of Ebolavirus in the domestic ferret, using wild-type nonadapted viruses. Pathologic features were consistent with disease in primates. Of particular importance, this is the only known small-animal model developed for Bundibugyo and the only uniformly lethal animal model for Bundibugyo.


The Journal of Infectious Diseases | 2015

Modeling the Disease Course of Zaire ebolavirus Infection in the Outbred Guinea Pig

Robert W. Cross; Karla A. Fenton; Joan B. Geisbert; Chad E. Mire; Thomas W. Geisbert

BACKGROUND Rodent models that accurately reflect human filovirus infection are needed as early screens for medical countermeasures. Prior work in rodents with the Zaire species of Ebola virus (ZEBOV) primarily used inbred mice and guinea pigs to model disease. However, these inbred species do not show some of the important features of primate ZEBOV infection, most notably, coagulation abnormalities. METHODS Thirty-six outbred guinea pigs were infected with guinea pig-adapted ZEBOV and examined sequentially over an 8-day period to investigate the pathologic events that lead to death. RESULTS Features of disease in ZEBOV-infected outbred guinea pigs were largely consistent with disease in humans and nonhuman primates and included early infection of macrophages and dendritiform cells, apoptosis of bystander lymphocytes, and increases in levels of proinflammatory cytokines. Most importantly, dysregulation of circulating levels of fibrinogen, protein C activity, and antifibrinolytic proteins and deposition of fibrin in tissues demonstrated both biochemical and microscopic evidence of disseminated intravascular coagulation. CONCLUSIONS These findings suggest that the outbred guinea pig model recapitulates ZEBOV infection of primates better than inbred rodent models, is useful for dissecting key events in the pathogenesis of ZEBOV, and is useful for evaluating candidate interventions prior to assessment in primates.


The Journal of Infectious Diseases | 2015

Lack of Marburg Virus Transmission From Experimentally Infected to Susceptible In-Contact Egyptian Fruit Bats

Janusz T. Paweska; Petrus Jansen van Vuren; Karla A. Fenton; Kerry Graves; Antoinette A. Grobbelaar; Naazneen Moolla; Patricia A. Leman; Jacqueline Weyer; Nadia Storm; Stewart D. McCulloch; Terence Peter Scott; Wanda Markotter; Lieza Odendaal; Sarah J. Clift; Thomas W. Geisbert; Martin Hale; Alan Kemp

Egyptian fruit bats (Rousettus aegyptiacus) were inoculated subcutaneously (n = 22) with Marburg virus (MARV). No deaths, overt signs of morbidity, or gross lesions was identified, but microscopic pathological changes were seen in the liver of infected bats. The virus was detected in 15 different tissues and plasma but only sporadically in mucosal swab samples, urine, and fecal samples. Neither seroconversion nor viremia could be demonstrated in any of the in-contact susceptible bats (n = 14) up to 42 days after exposure to infected bats. In bats rechallenged (n = 4) on day 48 after infection, there was no viremia, and the virus could not be isolated from any of the tissues tested. This study confirmed that infection profiles are consistent with MARV replication in a reservoir host but failed to demonstrate MARV transmission through direct physical contact or indirectly via air. Bats develop strong protective immunity after infection with MARV.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Monoclonal antibody therapy for Junin virus infection

Larry Zeitlin; Joan B. Geisbert; Daniel J. Deer; Karla A. Fenton; Ognian Bohorov; Natasha Bohorova; Charles Goodman; Do Han Kim; Andrew Hiatt; Michael Pauly; Jesus Velasco; Kevin J. Whaley; Friedrich Altmann; Clemens Gruber; Herta Steinkellner; Anna N. Honko; Ana I. Kuehne; M. Javad Aman; Sara Sahandi; Sven Enterlein; Xiaoguo Zhan; Delia A. Enria; Thomas W. Geisbert

Significance There are no Food and Drug Administration approved drugs available for preventing or treating Argentine hemorrhagic fever (AHF), and the current treatment option is limited to administration of immune plasma. With the expanding clinical use of monoclonal antibodies (mAbs) for acute and chronic conditions, it has become clear that mAbs offer a highly specific, potent, and generally safe drug platform for antivirals, and may be a useful alternative to immune plasma. Here, we show that mAbs are effective in the Junin virus guinea pig model and likely to be an economical therapy for AHF. Countermeasures against potential biothreat agents remain important to US Homeland Security, and many of these pharmaceuticals could have dual use in the improvement of global public health. Junin virus, the causative agent of Argentine hemorrhagic fever (AHF), is an arenavirus identified as a category A high-priority agent. There are no Food and Drug Administration (FDA) approved drugs available for preventing or treating AHF, and the current treatment option is limited to administration of immune plasma. Whereas immune plasma demonstrates the feasibility of passive immunotherapy, it is limited in quantity, variable in quality, and poses safety risks such as transmission of transfusion-borne diseases. In an effort to develop a monoclonal antibody (mAb)-based alternative to plasma, three previously described neutralizing murine mAbs were expressed as mouse-human chimeric antibodies and evaluated in the guinea pig model of AHF. These mAbs provided 100% protection against lethal challenge when administered 2 d after infection (dpi), and one of them (J199) was capable of providing 100% protection when treatment was initiated 6 dpi and 92% protection when initiated 7 dpi. The efficacy of J199 is superior to that previously described for all other evaluated drugs, and its high potency suggests that mAbs like J199 offer an economical alternative to immune plasma and an effective dual use (bioterrorism/public health) therapeutic.

Collaboration


Dive into the Karla A. Fenton's collaboration.

Top Co-Authors

Avatar

Thomas W. Geisbert

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Chad E. Mire

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Joan B. Geisbert

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Krystle N. Agans

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

Ian Maclachlan

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Benjamin A. Satterfield

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge