Network


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

Hotspot


Dive into the research topics where Kathleen A. Cashman is active.

Publication


Featured researches published by Kathleen A. Cashman.


Virology Journal | 2010

Lassa virus-like particles displaying all major immunological determinants as a vaccine candidate for Lassa hemorrhagic fever

Luis M. Branco; Jessica N Grove; Frederick J Geske; Matt L. Boisen; Ivana J Muncy; Susan Magliato; Lee A. Henderson; Randal J. Schoepp; Kathleen A. Cashman; Lisa E. Hensley; Robert F. Garry

BackgroundLassa fever is a neglected tropical disease with significant impact on the health care system, society, and economy of Western and Central African nations where it is endemic. Treatment of acute Lassa fever infections has successfully utilized intravenous administration of ribavirin, a nucleotide analogue drug, but this is not an approved use; efficacy of oral administration has not been demonstrated. To date, several potential new vaccine platforms have been explored, but none have progressed toward clinical trials and commercialization. Therefore, the development of a robust vaccine platform that could be generated in sufficient quantities and at a low cost per dose could herald a subcontinent-wide vaccination program. This would move Lassa endemic areas toward the control and reduction of major outbreaks and endemic infections. To this end, we have employed efficient mammalian expression systems to generate a Lassa virus (LASV)-like particle (VLP)-based modular vaccine platform.ResultsA mammalian expression system that generated large quantities of LASV VLP in human cells at small scale settings was developed. These VLP contained the major immunological determinants of the virus: glycoprotein complex, nucleoprotein, and Z matrix protein, with known post-translational modifications. The viral proteins packaged into LASV VLP were characterized, including glycosylation profiles of glycoprotein subunits GP1 and GP2, and structural compartmentalization of each polypeptide. The host cell protein component of LASV VLP was also partially analyzed, namely glycoprotein incorporation, though the identity of these proteins remain unknown. All combinations of LASV Z, GPC, and NP proteins that generated VLP did not incorporate host cell ribosomes, a known component of native arenaviral particles, despite detection of small RNA species packaged into pseudoparticles. Although VLP did not contain the same host cell components as the native virion, electron microscopy analysis demonstrated that LASV VLP appeared structurally similar to native virions, with pleiomorphic distribution in size and shape. LASV VLP that displayed GPC or GPC+NP were immunogenic in mice, and generated a significant IgG response to individual viral proteins over the course of three immunizations, in the absence of adjuvants. Furthermore, sera from convalescent Lassa fever patients recognized VLP in ELISA format, thus affirming the presence of native epitopes displayed by the recombinant pseudoparticles.ConclusionsThese results established that modular LASV VLP can be generated displaying high levels of immunogenic viral proteins, and that small laboratory scale mammalian expression systems are capable of producing multi-milligram quantities of pseudoparticles. These VLP are structurally and morphologically similar to native LASV virions, but lack replicative functions, and thus can be safely generated in low biosafety level settings. LASV VLP were immunogenic in mice in the absence of adjuvants, with mature IgG responses developing within a few weeks after the first immunization. These studies highlight the relevance of a VLP platform for designing an optimal vaccine candidate against Lassa hemorrhagic fever, and warrant further investigation in lethal challenge animal models to establish their protective potential.


Antiviral Research | 2011

Evaluation of Lassa antiviral compound ST-193 in a guinea pig model.

Kathleen A. Cashman; Mark A. Smith; Nancy A. Twenhafel; Ryan A. Larson; Kevin F. Jones; Robert D. Allen; Dongcheng Dai; Jarasvech Chinsangaram; Tove' C. Bolken; Dennis E. Hruby; Sean M. Amberg; Lisa E. Hensley; Mary C. Guttieri

Abstract Lassa virus (LASV), a member of the Arenaviridae family, causes a viral hemorrhagic fever endemic to West Africa, where as many as 300,000 infections occur per year. Presently, there are no FDA-approved LASV-specific vaccines or antiviral agents, although the antiviral drug ribavirin has shown some efficacy. A recently identified small-molecule inhibitor of arenavirus entry, ST-193, exhibits submicromolar antiviral activity in vitro. To determine the antiviral utility of ST-193 in vivo, we tested the efficacy of this compound in the LASV guinea pig model. Four groups of strain 13 guinea pigs were administered 25 or 80mg/kg ST-193, 25mg/kg of ribavirin, or the vehicle by the intraperitoneal (i.p.) route before infection with a lethal dose of LASV, strain Josiah, and continuing once daily for 14 days. Control animals exhibited severe disease, becoming moribund between days 10 and 15 postinfection. ST-193-treated animals exhibited fewer signs of disease and enhanced survival when compared to the ribavirin or vehicle groups. Body temperatures in all groups were elevated by day 9, but returned to normal by day 19 postinfection in the majority of ST-193-treated animals. ST-193 treatment mediated a 2–3-log reduction in viremia relative to vehicle-treated controls. The overall survival rate for the ST-193-treated guinea pigs was 62.5% (10/16) compared with 0% in the ribavirin (0/8) and vehicle (0/7) groups. These data suggest that ST-193 may serve as an improved candidate for the treatment of Lassa fever.


Nature microbiology | 2017

Identification and pathological characterization of persistent asymptomatic Ebola virus infection in rhesus monkeys

Xiankun Zeng; Candace D. Blancett; Keith A Koistinen; Christopher W. Schellhase; Jeremy J. Bearss; Sheli R. Radoshitzky; Shelley P. Honnold; Taylor B. Chance; Travis K. Warren; Jeffrey W. Froude; Kathleen A. Cashman; John M. Dye; Sina Bavari; Gustavo Palacios; Jens H. Kuhn; Mei G. Sun

Ebola virus (EBOV) persistence in asymptomatic humans and Ebola virus disease (EVD) sequelae have emerged as significant public health concerns since the 2013–2016 EVD outbreak in Western Africa. Until now, studying how EBOV disseminates into and persists in immune-privileged sites was impossible due to the absence of a suitable animal model. Here, we detect persistent EBOV replication coinciding with systematic inflammatory responses in otherwise asymptomatic rhesus monkeys that had survived infection in the absence of or after treatment with candidate medical countermeasures. We document progressive EBOV dissemination into the eyes, brain and testes through vascular structures, similar to observations in humans. We identify CD68+ cells (macrophages/monocytes) as the cryptic EBOV reservoir cells in the vitreous humour and its immediately adjacent tissue, in the tubular lumina of the epididymides, and in foci of histiocytic inflammation in the brain, but not in organs typically affected during acute infection. In conclusion, our data suggest that persistent EBOV infection in rhesus monkeys could serve as a model for persistent EBOV infection in humans, and we demonstrate that promising candidate medical countermeasures may not completely clear EBOV infection. A rhesus monkey model may lay the foundation to study EVD sequelae and to develop therapies to abolish EBOV persistence.


Virology Journal | 2008

Bacterial-based systems for expression and purification of recombinant Lassa virus proteins of immunological relevance.

Luis M. Branco; Alex Matschiner; Joseph N. Fair; Augustine Goba; Darryl B. Sampey; Philip J Ferro; Kathleen A. Cashman; Randal J. Schoepp; Robert B. Tesh; Daniel G. Bausch; Robert F. Garry; Mary C. Guttieri

BackgroundThere is a significant requirement for the development and acquisition of reagents that will facilitate effective diagnosis, treatment, and prevention of Lassa fever. In this regard, recombinant Lassa virus (LASV) proteins may serve as valuable tools in diverse antiviral applications. Bacterial-based systems were engineered for expression and purification of recombinant LASV nucleoprotein (NP), glycoprotein 1 (GP1), and glycoprotein 2 (GP2).ResultsFull-length NP and the ectodomains of GP1 and GP2 were generated as maltose-binding protein (MBP) fusions in the Rosetta strains of Escherichia coli (E. coli) using pMAL-c2x vectors. Average fusion protein yields per liter of culture for MBP-NP, MBP-GP1, and MBP-GP2 were 10 mg, 9 mg, and 9 mg, respectively. Each protein was captured from cell lysates using amylose resin, cleaved with Factor Xa, and purified using size-exclusion chromatography (SEC). Fermentation cultures resulted in average yields per liter of 1.6 mg, 1.5 mg, and 0.7 mg of purified NP, GP1 and GP2, respectively. LASV-specific antibodies in human convalescent sera specifically detected each of the purified recombinant LASV proteins, highlighting their utility in diagnostic applications. In addition, mouse hyperimmune ascitic fluids (MHAF) against a panel of Old and New World arenaviruses demonstrated selective cross reactivity with LASV proteins in Western blot and enzyme-linked immunosorbent assay (ELISA).ConclusionThese results demonstrate the potential for developing broadly reactive immunological assays that employ all three arenaviral proteins individually and in combination.


Vaccine | 2013

Enhanced Efficacy of a Codon-Optimized DNA Vaccine Encoding the Glycoprotein Precursor Gene of Lassa Virus in a Guinea Pig Disease Model When Delivered by Dermal Electroporation

Kathleen A. Cashman; Kate E. Broderick; Eric R. Wilkinson; Carl I. Shaia; Todd M. Bell; Amy C. Shurtleff; Kristin Spik; Catherine V. Badger; Mary C. Guttieri; Niranjan Y. Sardesai; Connie S. Schmaljohn

Lassa virus (LASV) causes a severe, often fatal, hemorrhagic fever endemic to West Africa. Presently, there are no FDA-licensed medical countermeasures for this disease. In a pilot study, we constructed a DNA vaccine (pLASV-GPC) that expressed the LASV glycoprotein precursor gene (GPC). This plasmid was used to vaccinate guinea pigs (GPs) using intramuscular electroporation as the delivery platform. Vaccinated GPs were protected from lethal infection (5/6) with LASV compared to the controls. However, vaccinated GPs experienced transient viremia after challenge, although lower than the mock-vaccinated controls. In a follow-on study, we developed a new device that allowed for both the vaccine and electroporation pulse to be delivered to the dermis. We also codon-optimized the GPC sequence of the vaccine to enhance expression in GPs. Together, these innovations resulted in enhanced efficacy of the vaccine. Unlike the pilot study where neutralizing titers were not detected until after virus challenge, modest neutralizing titers were detected in guinea pigs before challenge, with escalating titers detected after challenge. The vaccinated GPs were never ill and were not viremic at any timepoint. The combination of the codon-optimized vaccine and dermal electroporation delivery is a worthy candidate for further development.


Human Vaccines & Immunotherapeutics | 2017

A DNA vaccine delivered by dermal electroporation fully protects cynomolgus macaques against Lassa fever

Kathleen A. Cashman; Eric R. Wilkinson; Carl I. Shaia; Paul R Facemire; Todd M. Bell; Jeremy J. Bearss; Joshua D. Shamblin; Suzanne E. Wollen; Kate E. Broderick; Niranjan Y. Sardesai; Connie S. Schmaljohn

ABSTRACT Lassa virus (LASV) is an ambisense RNA virus in the Arenaviridae family and is the etiological agent of Lassa fever, a severe hemorrhagic disease endemic to West and Central Africa.1,2 There are no US Food and Drug Administration (FDA)-licensed vaccines available to prevent Lassa fever.1,2 in our previous studies, we developed a gene-optimized DNA vaccine that encodes the glycoprotein precursor gene of LASV (Josiah strain) and demonstrated that 3 vaccinations accompanied by dermal electroporation protected guinea pigs from LASV-associated illness and death. Here, we describe an initial efficacy experiment in cynomolgus macaque nonhuman primates (NHPs) in which we followed an identical 3-dose vaccine schedule that was successful in guinea pigs, and a follow-on experiment in which we used an accelerated vaccination strategy consisting of 2 administrations, spaced 4 weeks apart. In both studies, all of the LASV DNA-vaccinated NHPs survived challenge and none of them had measureable, sustained viremia or displayed weight loss or other disease signs post-exposure. Three of 10 mock-vaccinates survived exposure to LASV, but all of them became acutely ill post-exposure and remained chronically ill to the study end point (45 d post-exposure). Two of the 3 survivors experienced sensorineural hearing loss (described elsewhere). These results clearly demonstrate that the LASV DNA vaccine combined with dermal electroporation is a highly effective candidate for eventual use in humans.


Human Vaccines & Immunotherapeutics | 2017

DNA vaccines elicit durable protective immunity against individual or simultaneous infections with Lassa and Ebola viruses in guinea pigs

Kathleen A. Cashman; Eric R. Wilkinson; Suzanne E. Wollen; Joshua D. Shamblin; Justine M. Zelko; Jeremy J. Bearss; Xiankun Zeng; Kate E. Broderick; Connie S. Schmaljohn

ABSTRACT We previously developed optimized DNA vaccines against both Lassa fever and Ebola hemorrhagic fever viruses and demonstrated that they were protective individually in guinea pig and nonhuman primate models. In this study, we vaccinated groups of strain 13 guinea pigs two times, four weeks apart with 50 µg of each DNA vaccine or a mock vaccine at discrete sites by intradermal electroporation. Five weeks following the second vaccinations, guinea pigs were exposed to lethal doses of Lassa virus, Ebola virus, or a combination of both viruses simultaneously. None of the vaccinated guinea pigs, regardless of challenge virus and including the coinfected group, displayed weight loss, fever or other disease signs, and all survived to the study endpoint. All of the mock-vaccinated guinea pigs that were infected with Lassa virus, and all but one of the EBOV-infected mock-vaccinated guinea pigs succumbed. In order to determine if the dual-agent vaccination strategy could protect against both viruses if exposures were temporally separated, we held the surviving vaccinates in BSL-4 for approximately 120 days to perform a cross-challenge experiment in which guinea pigs originally infected with Lassa virus received a lethal dose of Ebola virus and those originally infected with Ebola virus were infected with a lethal dose of Lassa virus. All guinea pigs remained healthy and survived to the study endpoint. This study clearly demonstrates that DNA vaccines against Lassa and Ebola viruses can elicit protective immunity against both individual virus exposures as well as in a mixed-infection environment.


Archive | 2012

Cross-protective arenavirus vaccines and their method of use

Kate E. Broderick; Niranjan Y. Sardesai; Kathleen A. Cashman


Archive | 2017

Autoimmune Associated Systemic Vasculitis as the Cause of Sudden onset Bilateral Sensorineural Hearing Loss Following Lassa Virus Exposure in a Cynomolgus Macaque Deafness Model

Kathleen A. Cashman; Eric R. Wilkinson; Paul R Facemire; Todd M. Bell; Carl I. Shaia; Connie S. Schmaljohn


Archive | 2016

Systemic Polyarteritis Nodosa as the Cause of Sudden Onset Bilateral Sensorineural Hearing Loss Following Lassa Virus Infection

Kathleen A. Cashman; Eric R. Wilkinson; Paul R Facemire; Todd M. Bell; Carl I. Shaia; Connie S. Schmaljohn

Collaboration


Dive into the Kathleen A. Cashman's collaboration.

Top Co-Authors

Avatar

Connie S. Schmaljohn

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Eric R. Wilkinson

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carl I. Shaia

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Mary C. Guttieri

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Todd M. Bell

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul R Facemire

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge