Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Heart Lung Transplant ; 41(3): 287-297, 2022 03.
Article in English | MEDLINE | ID: mdl-34802874

ABSTRACT

BACKGROUND: Transmission of latent human cytomegalovirus (HCMV) via organ transplantation with post-transplant viral reactivation is extremely prevalent and results in substantial adverse impact on outcomes. Therapies targeting the latent reservoir within the allograft to mitigate viral transmission would represent a major advance. Here, we delivered an immunotoxin (F49A-FTP) that targets and kills latent HCMV aiming at reducing the HCMV reservoir from donor lungs using ex-vivo lung perfusion (EVLP). METHODS: HCMV seropositive human lungs were placed on EVLP alone or EVLP + 1mg/L of F49A-FTP for 6 hours (n = 6, each). CD14+ monocytes isolated from biopsies pre and post EVLP underwent HCMV reactivation assay designed to evaluate viral reactivation capacity. Off-target effects of F49A-FTP were studied evaluating cell death markers of CD34+ and CD14+ cells using flow cytometry. Lung function on EVLP and inflammatory cytokine production were evaluated as safety endpoints. RESULTS: We demonstrate that lungs treated ex-vivo with F49A-FTP had a significant reduction in HCMV reactivation compared to controls, suggesting successful targeting of latent virus (76% median reduction in F49A-FTP vs 15% increase in controls, p = 0.0087). Furthermore, there was comparable cell death rates of the targeted cells between both groups, suggesting no off-target effects. Ex-vivo lung function was stable over 6 hours and no differences in key inflammatory cytokines were observed demonstrating safety of this novel treatment. CONCLUSIONS: Ex-vivo F49A-FTP treatment of human lungs targets and kills latent HCMV, markedly attenuating HCMV reactivation. This approach demonstrates the first experiments targeting latent HCMV in a donor organ with promising results towards clinical translation.


Subject(s)
Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Immunotoxins/pharmacology , Immunotoxins/therapeutic use , Lung Transplantation , Patient Selection , Chemokine CX3CL1 , Exotoxins , Humans , In Vitro Techniques
2.
J Immunol Res ; 2017: 4069260, 2017.
Article in English | MEDLINE | ID: mdl-28251165

ABSTRACT

Immunotoxins as antiviral therapeutics are largely unexplored but have promising prospective due to their high selectivity potential and their unparalleled efficiency. One recent example targeted the virus-encoded G protein-coupled receptor US28 as a strategy for specific and efficient treatment of human cytomegalovirus (HCMV) infections. US28 is expressed on virus-infected cells and scavenge chemokines by rapid internalization. The chemokine-based fusion-toxin protein (FTP) consisted of a variant (F49A) of CX3CL1 specifically targeting US28 linked to the catalytic domain of Pseudomonas exotoxin A (PE). Here, we systematically seek to improve F49A-FTP by modifications in its three structural domains; we generated variants with (1) altered chemokine sequence (K14A, F49L, and F49E), (2) shortened and elongated linker region, and (3) modified toxin domain. Only F49L-FTP displayed higher selectivity in its binding to US28 versus CX3CR1, the endogenous receptor for CX3CL1, but this was not matched by a more selective killing of US28-expressing cells. A longer linker and different toxin variants decreased US28 affinity and selective killing. Thereby, F49A-FTP represents the best candidate for HCMV treatment. Many viruses encode internalizing receptors suggesting that not only HCMV but also, for instance, Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus may be targeted by FTPs.


Subject(s)
Antiviral Agents/immunology , Antiviral Agents/therapeutic use , Cytomegalovirus/immunology , Immunotoxins/therapeutic use , Antiviral Agents/adverse effects , Bacterial Proteins/immunology , Cells, Cultured , Chemokine CX3CL1/metabolism , Chemokine CX3CL1/therapeutic use , Fibroblasts , Herpesvirus 4, Human , Humans , Immunotoxins/adverse effects , Prospective Studies , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Signal Transduction , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 112(27): 8427-32, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26080445

ABSTRACT

The use of receptor-ligand interactions to direct toxins to kill diseased cells selectively has shown considerable promise for treatment of a number of cancers and, more recently, autoimmune disease. Here we move the fusion toxin protein (FTP) technology beyond cancer/autoimmune therapeutics to target the human viral pathogen, human cytomegalovirus (HCMV), on the basis of its expression of the 7TM G protein-coupled chemokine receptor US28. The virus origin of US28 provides an exceptional chemokine-binding profile with high selectivity and improved binding for the CX3C chemokine, CX3CL1. Moreover, US28 is constitutively internalizing by nature, providing highly effective FTP delivery. We designed a synthetic CX3CL1 variant engineered to have ultra-high affinity for US28 and greater specificity for US28 than the natural sole receptor for CX3CL1, CX3CR1, and we fused the synthetic variant with the cytotoxic domain of Pseudomonas Exotoxin A. This novel strategy of a rationally designed FTP provided unparalleled anti-HCMV efficacy and potency in vitro and in vivo.


Subject(s)
Bacterial Proteins/metabolism , Chemokine CX3CL1/metabolism , Cytomegalovirus Infections/prevention & control , Receptors, Chemokine/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Viral Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Cell Line , Cell Survival/drug effects , Chemokine CX3CL1/genetics , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , Dose-Response Relationship, Drug , Drug Design , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/virology , HEK293 Cells , Host-Pathogen Interactions/drug effects , Humans , Lung/cytology , Protein Binding/drug effects , Receptors, Chemokine/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Time Factors , Viral Proteins/metabolism
4.
Anal Chem ; 80(10): 3648-54, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18422341

ABSTRACT

This manuscript presents, for the first time, the method of automated structural analysis of biomolecules in solution on a microfluidic chip. A polymer-based micrototal analysis system for high-throughput Small-Angle X-ray Scattering (SAXS) data collection from biological macromolecules has been developed. The bioXTAS chip features an integrated X-ray transparent 200 nL sample chamber and diffusion-based mixing of protein and buffer solutions. Software for fully automated fluidic control, data acquisition, and data analysis has been developed. The proof-of concept is based on data using bovine serum albumin as the model system. It confirms the quality of SAXS data generated from small sample volumes and furthermore validates the on-chip mixing capabilities. SAXS data on the gradual unfolding of BSA induced by an anionic surfactant exemplifies how the bioXTAS chip can be used to follow and identify structural changes and proves the feasibility of high-throughput structural analysis in solution. In total, this shows that the bioXTAS chip has the potential for becoming a powerful tool for automated high-throughput structural analysis of macromolecular systems.


Subject(s)
Microfluidics/instrumentation , Proteins/chemistry , Scattering, Radiation , Sodium Dodecyl Sulfate , Solutions
SELECTION OF CITATIONS
SEARCH DETAIL
...