Network


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

Hotspot


Dive into the research topics where Ricardo Carrion is active.

Publication


Featured researches published by Ricardo Carrion.


PLOS ONE | 2013

A Systematic Screen of FDA-Approved Drugs for Inhibitors of Biological Threat Agents

Peter B. Madrid; Sidharth Chopra; Ian D. Manger; Lynne Gilfillan; Tiffany R. Keepers; Amy C. Shurtleff; Carol E. Green; Lalitha V. Iyer; Holli Hutcheson Dilks; Robert A. Davey; Andrey A. Kolokoltsov; Ricardo Carrion; Jean L. Patterson; Sina Bavari; Rekha G. Panchal; Travis K. Warren; Jay Wells; Walter H. Moos; RaeLyn L. Burke; Mary J. Tanga

Background The rapid development of effective medical countermeasures against potential biological threat agents is vital. Repurposing existing drugs that may have unanticipated activities as potential countermeasures is one way to meet this important goal, since currently approved drugs already have well-established safety and pharmacokinetic profiles in patients, as well as manufacturing and distribution networks. Therefore, approved drugs could rapidly be made available for a new indication in an emergency. Methodology/Principal Findings A large systematic effort to determine whether existing drugs can be used against high containment bacterial and viral pathogens is described. We assembled and screened 1012 FDA-approved drugs for off-label broad-spectrum efficacy against Bacillus anthracis; Francisella tularensis; Coxiella burnetii; and Ebola, Marburg, and Lassa fever viruses using in vitro cell culture assays. We found a variety of hits against two or more of these biological threat pathogens, which were validated in secondary assays. As expected, antibiotic compounds were highly active against bacterial agents, but we did not identify any non-antibiotic compounds with broad-spectrum antibacterial activity. Lomefloxacin and erythromycin were found to be the most potent compounds in vivo protecting mice against Bacillus anthracis challenge. While multiple virus-specific inhibitors were identified, the most noteworthy antiviral compound identified was chloroquine, which disrupted entry and replication of two or more viruses in vitro and protected mice against Ebola virus challenge in vivo. Conclusions/Significance The feasibility of repurposing existing drugs to face novel threats is demonstrated and this represents the first effort to apply this approach to high containment bacteria and viruses.


Clinical and Vaccine Immunology | 2006

Detection of Anthrax Toxin in the Serum of Animals Infected with Bacillus anthracis by Using Engineered Immunoassays

Robert Mabry; Kathleen M. Brasky; Robert Geiger; Ricardo Carrion; Gene B. Hubbard; Stephen H. Leppla; Jean L. Patterson; George Georgiou; Brent L. Iverson

ABSTRACT Several strategies that target anthrax toxin are being developed as therapies for infection by Bacillus anthracis. Although the action of the tripartite anthrax toxin has been extensively studied in vitro, relatively little is known about the presence of toxins during an infection in vivo. We developed a series of sensitive sandwich enzyme-linked immunosorbent assays (ELISAs) for detection of both the protective antigen (PA) and lethal factor (LF) components of the anthrax exotoxin in serum. The assays utilize as capture agents an engineered high-affinity antibody to PA, a soluble form of the extracellular domain of the anthrax toxin receptor (ANTXR2/CMG2), or PA itself. Sandwich immunoassays were used to detect and quantify PA and LF in animals infected with the Ames or Vollum strains of anthrax spores. PA and LF were detected before and after signs of toxemia were observed, with increasing levels reported in the late stages of the infection. These results represent the detection of free PA and LF by ELISA in the systemic circulation of two animal models exposed to either of the two fully virulent strains of anthrax. Simple anthrax toxin detection ELISAs could prove useful in the evaluation of potential therapies and possibly as a clinical diagnostic to complement other strategies for the rapid identification of B. anthracis infection.


Journal of Virology | 2005

A live attenuated vaccine for Lassa fever made by reassortment of Lassa and Mopeia viruses.

Igor S. Lukashevich; Jean L. Patterson; Ricardo Carrion; Dmitry Moshkoff; Anysha Ticer; Juan Carlos Zapata; Kathleen M. Brasky; Robert Geiger; Gene B. Hubbard; Joseph Bryant; Maria S. Salvato

ABSTRACT Lassa virus (LASV) and Mopeia virus (MOPV) are closely related Old World arenaviruses that can exchange genomic segments (reassort) during coinfection. Clone ML29, selected from a library of MOPV/LASV (MOP/LAS) reassortants, encodes the major antigens (nucleocapsid and glycoprotein) of LASV and the RNA polymerase and zinc-binding protein of MOPV. Replication of ML29 was attenuated in guinea pigs and nonhuman primates. In murine adoptive-transfer experiments, as little as 150 PFU of ML29 induced protective cell-mediated immunity. All strain 13 guinea pigs vaccinated with clone ML29 survived at least 70 days after LASV challenge without either disease signs or histological lesions. Rhesus macaques inoculated with clone ML29 developed primary virus-specific T cells capable of secreting gamma interferon in response to homologous MOP/LAS and heterologous MOPV and lymphocytic choriomeningitis virus. Detailed examination of two rhesus macaques infected with this MOPV/LAS reassortant revealed no histological lesions or disease signs. Thus, ML29 is a promising attenuated vaccine candidate for Lassa fever.


Virology | 2009

Protection against lethal challenge by Ebola virus-like particles produced in insect cells

Yuliang Sun; Ricardo Carrion; Ling Ye; Zhiyuan Wen; Young Tae Ro; Kathleen M. Brasky; Anysha Ticer; E. Ellen Schwegler; Jean L. Patterson; Richard W. Compans; Chinglai Yang

Ebola virus-like particles (VLPs) were produced in insect cells using a recombinant baculovirus expression system and their efficacy for protection against Ebola virus infection was investigated. Two immunizations with 50 microg Ebola VLPs (high dose) induced a high level of antibodies against Ebola GP that exhibited strong neutralizing activity against GP-mediated virus infection and conferred complete protection of vaccinated mice against lethal challenge by a high dose of mouse-adapted Ebola virus. In contrast, two immunizations with 10 microg Ebola VLPs (low dose) induced 5-fold lower levels of antibodies against GP and these mice were not protected against lethal Ebola virus challenge, similar to control mice that were immunized with 50 microg SIV Gag VLPs. However, the antibody responses against GP were boosted significantly after a third immunization with 10 microg Ebola VLPs to similar levels as those induced by two immunizations with 50 microg Ebola VLPs, and vaccinated mice were also effectively protected against lethal Ebola virus challenge. Furthermore, serum viremia levels in protected mice were either below the level of detection or significantly lower compared to the viremia levels in control mice. These results show that effective protection can be achieved by immunization with Ebola VLPs produced in insect cells, which give high production yields, and lend further support to their development as an effective vaccine strategy against Ebola virus.


Journal of Virology | 2007

Lassa Virus Infection in Experimentally Infected Marmosets: Liver Pathology and Immunophenotypic Alterations in Target Tissues

Ricardo Carrion; Kathleen M. Brasky; Keith G. Mansfield; Curtis Johnson; Monica Gonzales; Anysha Ticer; Igor S. Lukashevich; Suzette D. Tardif; Jean L. Patterson

ABSTRACT Lassa virus causes thousands of deaths annually in western Africa and is considered a potential biological weapon. In an attempt to develop a small nonhuman primate model of Lassa fever, common marmosets were subcutaneously inoculated with Lassa virus strain Josiah. This inoculation resulted in a systemic disease with clinical and morphological features mirroring those in fatal human Lassa infection: fever, weight loss, high viremia and viral RNA load in tissues, elevated liver enzymes, and severe morbidity between days 15 and 20. The most prominent histopathology findings included multifocal hepatic necrosis with mild inflammation and hepatocyte proliferation, lymphoid depletion, and interstitial nephritis. Cellular aggregates in regions of hepatocellular necrosis were largely composed of HAM56-positive macrophages, devoid of CD3-positive and CD20-positive cells, and characterized by marked reductions in the intensity of HLA-DP, DQ, DR staining. A marked reduction in the major histocompatibility complex class II expression was also observed in the lymph nodes. Immunophenotypic alterations in spleen included reductions in overall numbers of CD20-positive and CD3-positive cells and the disruption of lymphoid follicular architecture. These findings identify the common marmoset as an appropriate model of human Lassa fever and present the first experimental evidence that replication of Lassa virus in tissues is associated with alterations that would be expected to impair adaptive immunity.


Vaccine | 2008

Safety, immunogenicity, and efficacy of the ML29 reassortant vaccine for Lassa fever in small non-human primates

Igor S. Lukashevich; Ricardo Carrion; Maria S. Salvato; Keith G. Mansfield; Kathleen M. Brasky; Juan Carlos Zapata; Cristiana Cairo; Marco Goicochea; Gia E. Hoosien; Anysha Ticer; Joseph Bryant; Harry Davis; Rasha Hammamieh; Maria Mayda; Marti Jett; Jean L. Patterson

A single injection of ML29 reassortant vaccine for Lassa fever induces low, transient viremia, and low or moderate levels of ML29 replication in tissues of common marmosets depending on the dose of the vaccination. The vaccination elicits specific immune responses and completely protects marmosets against fatal disease by induction of sterilizing cell-mediated immunity. DNA array analysis of human peripheral blood mononuclear cells from healthy donors exposed to ML29 revealed that gene expression patterns in ML29-exposed PBMC and control, media-exposed PBMC, clustered together confirming safety profile of the ML29 in non-human primates. The ML29 reassortant is a promising vaccine candidate for Lassa fever.


Science Translational Medicine | 2012

Productive Replication of Ebola Virus Is Regulated by the c-Abl1 Tyrosine Kinase

Mayra García; Arik Cooper; Wei Shi; William G. Bornmann; Ricardo Carrion; Daniel Kalman; Gary J. Nabel

Ebola virus growth is regulated by a c-Abl tyrosine kinase, which modulates the viral protein VP40, and can be blocked by c-Abl antagonists. Disabling Ebola virus Ebola virus causes severe, often fatal, hemorrhagic fever in humans and is well known for its fulminant replication that ultimately overwhelms the capacity of the human immune system to contain it. Because it is easily transmitted from one human to another and causes high mortality, Ebola virus has been classified as a category A agent, the highest-priority biological threat classification. There are no effective prophylactic or therapeutic interventions available to treat infections with Ebola virus or another highly pathogenic filovirus, Marburg virus. Information about mechanisms of assembly and release of these viruses will be crucial for developing effective countermeasures. Viruses exploit a variety of components within the host cell to complete their life cycles. In a new study, García et al. show that the growth of Ebola virus is regulated by the c-Abl1 tyrosine kinase. They demonstrate that release of Ebola virus–like particles in a cell culture cotransfection system could be blocked by c-Abl1–specific small interfering RNAs (siRNAs) or by Abl-specific kinase inhibitors such as nilotinib. They show that this effect is mediated by tyrosine phosphorylation of the Ebola matrix protein VP40. The researchers identified at least one site of tyrosine phosphorylation (Y13) on VP40 and demonstrated that mutation of this residue reduced release of Ebola virus–like particles. The drug nilotinib or siRNAs directed against c-Abl1 also reduced productive replication of the Zaire strain of Ebola virus in cultured Vero cells by up to four orders of magnitude. Together, these data indicate that the c-Abl1 tyrosine kinase regulates budding or release of Ebola virus through a mechanism involving phosphorylation of VP40. The authors propose that this critical interaction between a viral and a host cell protein represents a target for antiviral therapy. These findings suggest that drugs such as nilotinib that inhibit c-Abl1 and are already in clinical use as anticancer therapeutics may be useful for reducing the severity of Ebola virus infection. Ebola virus causes a fulminant infection in humans resulting in diffuse bleeding, vascular instability, hypotensive shock, and often death. Because of its high mortality and ease of transmission from human to human, Ebola virus remains a biological threat for which effective preventive and therapeutic interventions are needed. An understanding of the mechanisms of Ebola virus pathogenesis is critical for developing antiviral therapeutics. Here, we report that productive replication of Ebola virus is modulated by the c-Abl1 tyrosine kinase. Release of Ebola virus–like particles (VLPs) in a cell culture cotransfection system was inhibited by c-Abl1–specific small interfering RNA (siRNA) or by Abl-specific kinase inhibitors and required tyrosine phosphorylation of the Ebola matrix protein VP40. Expression of c-Abl1 stimulated an increase in phosphorylation of tyrosine 13 (Y13) of VP40, and mutation of Y13 to alanine decreased the release of Ebola VLPs. Productive replication of the highly pathogenic Ebola virus Zaire strain was inhibited by c-Abl1–specific siRNAs or by the Abl-family inhibitor nilotinib by up to four orders of magnitude. These data indicate that c-Abl1 regulates budding or release of filoviruses through a mechanism involving phosphorylation of VP40. This step of the virus life cycle therefore may represent a target for antiviral therapy.


Infection and Immunity | 2005

Passive protection against anthrax by using a high-affinity antitoxin antibody fragment lacking an Fc region.

Robert Mabry; Robert Geiger; Gene B. Hubbard; Ricardo Carrion; Kathleen M. Brasky; Jean L. Patterson; George Georgiou; Brent L. Iverson

ABSTRACT Passive immunization has been successfully employed for protection against bacterial and viral infections for over 100 years. Immunoglobulin Fc regions play a critical role in the clearance of bacterial pathogens by mediating antibody-dependent and complement-dependent cytotoxicity. Here we show that antibody fragments engineered to recognize the protective antigen component of the B. anthracis exotoxin with high affinity and conjugated to polyethylene glycol (PEG) for prolonged circulation half-life confer significant protection against inhalation anthrax despite their lack of Fc regions. The speed and lower manufacturing cost of bacterially expressed PEGylated antibody fragments could provide decisive advantages for anthrax prophylaxis. Importantly, our results suggest that PEGylated antibody fragments may represent a unique approach for mounting a rapid therapeutic response to emerging pathogen infections.


Clinical and Vaccine Immunology | 2009

Immunogenicity and Protective Efficacy of a Recombinant Subunit West Nile Virus Vaccine in Rhesus Monkeys

Michael M. Lieberman; Vivek R. Nerurkar; Haiyan Luo; Bruce Cropp; Ricardo Carrion; Melissa de la Garza; Beth Ann Coller; D. L. Clements; Steven A. Ogata; Teri Wong; Tim Martyak; Carolyn Weeks-Levy

ABSTRACT The immunogenicity and protective efficacy of a recombinant subunit West Nile virus (WNV) vaccine was evaluated in rhesus macaques (Macaca mulatta). The vaccine consisted of a recombinant envelope (E) protein truncated at the C-terminal end, resulting in a polypeptide containing 80% of the N-terminal amino acids of the native WNV protein (WN-80E), mixed with an adjuvant (GPI-0100). WN-80E was produced in a Drosophila melanogaster expression system with high yield and purified by immunoaffinity chromatography using a monoclonal antibody specific for flavivirus E proteins. Groups of monkeys were vaccinated with formulations containing 1 or 25 μg of WN-80E antigen, and both humoral and cellular immunity were assessed after vaccination. The results demonstrated potent antibody responses to vaccination, as determined by both enzyme-linked immunosorbent assay and virus-neutralizing antibody assays. All vaccinated animals responded favorably, and there was little difference in response between animals immunized with 1 or 25 μg of WN-80E. Cellular immunity was determined by lymphocyte proliferation and cytokine production assays using peripheral blood mononuclear cells from vaccinated animals stimulated in vitro with WN-80E. Cell-mediated immune responses varied from animal to animal within each group. About half of the animals responded with lymphoproliferation, cytokine production, or both. Again, there was little difference in response between animals immunized with a 1- or 25-μg dose of WN-80E in the vaccine formulations. In a separate experiment, groups of monkeys were immunized with the WN-80E/GPI-0100 vaccine or an adjuvant-only control formulation. Animals were then challenged by inoculation of wild-type WNV, and the level of viremia in each animal was monitored daily for 10 days. The results showed that whereas all animals in the control group had detectable viremia for at least 3 days after challenge, all of the vaccinated animals were negative on all days after challenge. Thus, the WN-80E vaccine was 100% efficacious in protecting monkeys against infection with WNV.


The Journal of Infectious Diseases | 2007

Rapid assembly of sensitive antigen-capture assays for Marburg virus, using in vitro selection of llama single-domain antibodies, at biosafety level 4.

Laura J. Sherwood; Lisa E. Osborn; Ricardo Carrion; Jean L. Patterson; Andrew Hayhurst

There is a pressing need for rapid and reliable approaches to the delivery of sensitive yet rugged diagnostic assays specific for emerging viruses, to hasten containment of outbreaks when and wherever they occur. Within 3 weeks, we delivered an antigen-capture assay for Marburg virus (MARV) that was based on llama single-domain antibodies (sdAbs) selected at biosafety level 4. Four unique sdAbs were capable of independently detecting MARV variants Musoke, Ravn, and Angola without cross-reactivity with the 4 Ebola virus species. The unoptimized assays could be performed in <30 min and, at best, provided a visual read of 10-100 pfu in a 100-microL sample when a colorimetric substrate was used and 0.1-1 pfu when a chemiluminescent substrate was used. All the sdAbs were specific for nucleoprotein, with an assay sensitivity that was reliant on detergent-mediated exposure of polyvalent antigen. Our strategy highlights the potential of direct antibody selection on filoviruses as a guide for effective and fast diagnostic development.

Collaboration


Dive into the Ricardo Carrion's collaboration.

Top Co-Authors

Avatar

Jean L. Patterson

Texas Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Kathleen M. Brasky

Texas Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Anysha Ticer

Texas Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Anthony Griffiths

Texas Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Holger Schmidt

University of California

View shared research outputs
Top Co-Authors

Avatar

Kendra J. Alfson

Texas Biomedical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge