Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Mol Pharm ; 20(1): 370-382, 2023 01 02.
Article in English | MEDLINE | ID: mdl-36484496

ABSTRACT

DNA viruses are responsible for many diseases in humans. Current treatments are often limited by toxicity, as in the case of cidofovir (CDV, Vistide), a compound used against cytomegalovirus (CMV) and adenovirus (AdV) infections. CDV is a polar molecule with poor bioavailability, and its overall clinical utility is limited by the high occurrence of acute nephrotoxicity. To circumvent these disadvantages, we designed nine CDV prodrug analogues. The prodrugs modulate the polarity of CDV with a long sulfonyl alkyl chain attached to one of the phosphono oxygens. We added capping groups to the end of the alkyl chain to minimize ß-oxidation and focus the metabolism on the phosphoester hydrolysis, thereby tuning the rate of this reaction by altering the alkyl chain length. With these modifications, the prodrugs have excellent aqueous solubility, optimized metabolic stability, increased cellular permeability, and rapid intracellular conversion to the pharmacologically active diphosphate form (CDV-PP). The prodrugs exhibited significantly enhanced antiviral potency against a wide range of DNA viruses in infected human foreskin fibroblasts. Single-dose intravenous and oral pharmacokinetic experiments showed that the compounds maintained plasma and target tissue levels of CDV well above the EC50 for 24 h. These experiments identified a novel lead candidate, NPP-669. NPP-669 demonstrated efficacy against CMV infections in mice and AdV infections in hamsters following oral (p.o.) dosing at a dose of 1 mg/kg BID and 0.1 mg/kg QD, respectively. We further showed that NPP-669 at 30 mg/kg QD did not exhibit histological signs of toxicity in mice or hamsters. These data suggest that NPP-669 is a promising lead candidate for a broad-spectrum antiviral compound.


Subject(s)
Cytomegalovirus Infections , Organophosphonates , Prodrugs , Mice , Humans , Animals , Antiviral Agents/pharmacokinetics , Biological Availability , Prodrugs/pharmacology , Cytosine , Cidofovir
2.
Antimicrob Agents Chemother ; 64(11)2020 10 20.
Article in English | MEDLINE | ID: mdl-32816736

ABSTRACT

Human adenovirus (HAdV) infection is common in the general population and can cause a range of clinical manifestations, among which pneumonia and keratoconjunctivitis are the most common. Although HAdV infections are mostly self-limiting, infections in immunocompromised individuals can be severe. No antiviral drug has been approved for treating adenoviruses. Filociclovir (FCV) is a nucleoside analogue which has successfully completed phase I human clinical safety studies and is now being developed for treatment of human cytomegalovirus (HCMV)-related disease in immunocompromised patients. In this report, we show that FCV is a potent broad-spectrum inhibitor of HAdV types 4 to 8, with 50% effective concentrations (EC50s) ranging between 1.24 and 3.6 µM and a 50% cytotoxic concentration (CC50) of 100 to 150 µM in human foreskin fibroblasts (HFFs). We also show that the prophylactic oral administration of FCV (10 mg/kg of body weight) 1 day prior to virus challenge and then daily for 14 days to immunosuppressed Syrian hamsters infected intravenously with HAdV6 was sufficient to prevent morbidity and mortality. FCV also mitigated tissue damage and inhibited virus replication in the liver. The 10-mg/kg dose had similar effects even when the treatment was started on day 4 after virus challenge. Furthermore, FCV administered at the same dose after intranasal challenge with HAdV6 partially mitigated body weight loss but significantly reduced pathology and virus replication in the lung. These findings suggest that FCV could potentially be developed as a pan-adenoviral inhibitor.


Subject(s)
Adenovirus Infections, Human , Adenoviruses, Human , Cytomegalovirus Infections , Adenovirus Infections, Human/drug therapy , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cricetinae , Cytomegalovirus Infections/drug therapy , Humans , Virus Replication
3.
J Virol ; 91(10)2017 05 15.
Article in English | MEDLINE | ID: mdl-28250128

ABSTRACT

Syrian hamsters are permissive for the replication of species C human adenoviruses (HAdV-C). The virus replicates to high titers in the liver of these animals after intravenous infection, while respiratory infection results in virus replication in the lung. Here we show that two types belonging to species C, HAdV-C5 and HAdV-C6, replicate to significantly different extents and cause pathology with significantly different severities, with HAdV-C6 replicating better and inducing more severe and more widespread lesions. The virus burdens in the livers of HAdV-C6-infected hamsters are higher than the virus burdens in HAdV-C5-infected ones because more of the permissive hepatocytes get infected. Furthermore, when hamsters are infected intravenously with HAdV-C6, live, infectious virus can be isolated from the lung and the kidney, which is not seen with HAdV-C5. Similarly to mouse models, in hamsters, HAdV-C6 is sequestered by macrophages to a lesser degree than HAdV-C5. Depletion of Kupffer cells from the liver greatly increases the replication of HAdV-C5 in the liver, while it has only a modest effect on the replication of HAdV-C6. Elimination of Kupffer cells also dramatically increases the pathology induced by HAdV-C5. These findings indicate that in hamsters, pathology resulting from intravenous infection with adenoviruses is caused mostly by replication in hepatocytes and not by the abortive infection of Kupffer cells and the following cytokine storm.IMPORTANCE Immunocompromised human patients can develop severe, often lethal adenovirus infections. Respiratory adenovirus infection among military recruits is a serious problem, in some cases requiring hospitalization of the patient. Furthermore, adenovirus-based vectors are frequently used as experimental viral therapeutic agents. Thus, it is imperative that we investigate the pathogenesis of adenoviruses in a permissive animal model. Syrian hamsters are susceptible to infection with certain human adenoviruses, and the pathology accompanying these infections is similar to what is observed with adenovirus-infected human patients. We demonstrate that replication in permissive cells in a susceptible host animal is a major part of the mechanism by which systemic adenovirus infection induces pathology, as opposed to the chiefly immune-mediated pathology observed in nonsusceptible hosts. These findings support the use of compounds inhibiting adenovirus replication as a means to block adenovirus-induced pathology.


Subject(s)
Adenovirus Infections, Human/pathology , Adenovirus Infections, Human/virology , Adenoviruses, Human/pathogenicity , Liver/virology , Viral Load , Virus Replication , Adenoviruses, Human/classification , Adenoviruses, Human/physiology , Animals , Cell Line , Cricetinae , Disease Models, Animal , Humans , Kidney/virology , Kupffer Cells/virology , Liver/pathology , Lung/virology , Macrophages/virology , Mesocricetus
5.
PLoS Pathog ; 11(8): e1005084, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26291525

ABSTRACT

Human adenoviruses have been studied extensively in cell culture and have been a model for studies in molecular, cellular, and medical biology. However, much less is known about adenovirus replication and pathogenesis in vivo in a permissive host because of the lack of an adequate animal model. Presently, the most frequently used permissive immunocompetent animal model for human adenovirus infection is the Syrian hamster. Species C human adenoviruses replicate in these animals and cause pathology that is similar to that seen with humans. Here, we report findings with a new Syrian hamster strain in which the STAT2 gene was functionally knocked out by site-specific gene targeting. Adenovirus-infected STAT2 knockout hamsters demonstrated an accentuated pathology compared to the wild-type control animals, and the virus load in the organs of STAT2 knockout animals was 100- to 1000-fold higher than that in wild-type hamsters. Notably, the adaptive immune response to adenovirus is not adversely affected in STAT2 knockout hamsters, and surviving hamsters cleared the infection by 7 to 10 days post challenge. We show that the Type I interferon pathway is disrupted in these hamsters, revealing the critical role of interferon-stimulated genes in controlling adenovirus infection. This is the first study to report findings with a genetically modified Syrian hamster infected with a virus. Further, this is the first study to show that the Type I interferon pathway plays a role in inhibiting human adenovirus replication in a permissive animal model. Besides providing an insight into adenovirus infection in humans, our results are also interesting from the perspective of the animal model: STAT2 knockout Syrian hamster may also be an important animal model for studying other viral infections, including Ebola-, hanta-, and dengue viruses, where Type I interferon-mediated innate immunity prevents wild type hamsters from being effectively infected to be used as animal models.


Subject(s)
Adenoviridae Infections/immunology , Adenoviruses, Human/pathogenicity , Disease Models, Animal , Interferon Type I/immunology , STAT2 Transcription Factor/deficiency , Adenoviridae Infections/pathology , Adenoviruses, Human/immunology , Animals , Animals, Genetically Modified , Cell Line, Tumor , Cricetinae , Flow Cytometry , Gene Knockout Techniques , Humans , Mesocricetus , Reverse Transcriptase Polymerase Chain Reaction , STAT2 Transcription Factor/immunology
6.
Antimicrob Agents Chemother ; 58(12): 7171-81, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224011

ABSTRACT

Adenovirus infections of immunocompromised patients can develop into deadly multiorgan or systemic disease. The virus is especially threatening for pediatric allogeneic hematopoietic stem cell transplant recipients; according to some studies, 10% or more of these patients succumb to disease resulting from adenovirus infection. At present, there is no drug approved for the treatment or prevention of adenovirus infections. Compounds that are approved to treat other virus infections are used off-label to combat adenovirus, but only anecdotal evidence of the efficacy of these drugs exists. Ganciclovir, a drug approved for the treatment of herpesvirus infection, was previously reported to be effective against human adenoviruses in vitro. To model adenovirus infections in immunocompromised humans, we examined ganciclovir's efficacy in immunosuppressed Syrian hamsters intravenously infected with type 5 human adenovirus (Ad5). This animal model is permissive for Ad5 replication, and the animals develop symptoms similar to those seen in humans. We demonstrate that ganciclovir suppresses Ad5 replication in the liver of infected hamsters and that it mitigates the consequences of Ad5 infections in these animals when administered prophylactically or therapeutically. We show that ganciclovir inhibits Ad5 DNA synthesis and late gene expression. The mechanism of action for the drug is not clear; preliminary data suggest that it exerts its antiadenoviral effect by directly inhibiting the adenoviral DNA polymerase. While more extensive studies are required, we believe that ganciclovir is a promising drug candidate to treat adenovirus infections. Brincidofovir, a drug with proven activity against Ad5, was used as a positive control in the prophylactic experiment.


Subject(s)
Adenoviridae Infections/drug therapy , Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Ganciclovir/pharmacology , Immunocompromised Host , Viral Proteins/antagonists & inhibitors , Adenoviridae Infections/immunology , Adenoviridae Infections/mortality , Adenoviridae Infections/virology , Adenoviruses, Human/genetics , Adenoviruses, Human/growth & development , Adenoviruses, Human/pathogenicity , Animals , Body Weight/drug effects , Cell Line, Tumor , Cytosine/analogs & derivatives , Cytosine/pharmacology , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Female , Gene Expression , Humans , Male , Mesocricetus , Organophosphonates/pharmacology , Survival Analysis , Transaminases/blood , Viral Load/drug effects , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
7.
Antimicrob Agents Chemother ; 58(12): 7451-61, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25267681

ABSTRACT

Herpesviruses are large double-stranded DNA viruses that cause serious human diseases. Herpesvirus DNA replication depends on multiple processes typically catalyzed by nucleotidyltransferase superfamily (NTS) enzymes. Therefore, we investigated whether inhibitors of NTS enzymes would suppress replication of herpes simplex virus 1 (HSV-1) and HSV-2. Eight of 42 NTS inhibitors suppressed HSV-1 and/or HSV-2 replication by >10-fold at 5 µM, with suppression at 50 µM reaching ∼1 million-fold. Five compounds in two chemical families inhibited HSV replication in Vero and human foreskin fibroblast cells as well as the approved drug acyclovir did. The compounds had 50% effective concentration values as low as 0.22 µM with negligible cytotoxicity in the assays employed. The inhibitors suppressed accumulation of viral genomes and infectious particles and blocked events in the viral replication cycle before and during viral DNA replication. Acyclovir-resistant mutants of HSV-1 and HSV-2 remained highly sensitive to the NTS inhibitors. Five of six NTS inhibitors of the HSVs also blocked replication of another herpesvirus pathogen, human cytomegalovirus. Therefore, NTS enzyme inhibitors are promising candidates for new herpesvirus treatments that may have broad efficacy against members of the herpesvirus family.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Nucleotidyltransferases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Viral Proteins/antagonists & inhibitors , Acyclovir/pharmacology , Animals , Chlorocebus aethiops , Cytomegalovirus/drug effects , Cytomegalovirus/enzymology , Cytomegalovirus/growth & development , DNA Replication/drug effects , DNA, Viral/antagonists & inhibitors , DNA, Viral/genetics , DNA, Viral/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Viral/drug effects , Fibroblasts/drug effects , Fibroblasts/virology , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/growth & development , Herpesvirus 2, Human/enzymology , Herpesvirus 2, Human/growth & development , Humans , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Primary Cell Culture , Vero Cells , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
8.
Antiviral Res ; 222: 105799, 2024 02.
Article in English | MEDLINE | ID: mdl-38190973

ABSTRACT

Adenovirus infections of immunocompromised humans are a significant source of morbidity and mortality. Presently, there is no drug specifically approved for the treatment of adenovirus infections by the FDA. The state-of-the-art treatment of such infections is the off-label use of cidofovir, an acyclic nucleotide phosphonate. While cidofovir inhibits adenovirus replication, it has dose-limiting kidney toxicity. There is an apparent need for a better compound to treat adenovirus infections. To this end, we have been developing acyclic nucleotide phosphonate prodrugs that utilize an amino acid scaffold equipped with a lipophilic modifier. Here, we compare the antiviral potential of two prodrugs of HPMPA that differ only in the amino acid-based promoiety: USC-087, based on an N-hexadecyl tyrosinamide, and USC-093, based on an N-hexadecyl serinamide. Oral administration of both compounds was very efficacious against disseminated HAdV-C6 infection in immunosuppressed Syrian hamsters, suppressing virus replication and mitigating pathology even when treatment was withheld until 4 days after challenge. We saw only marginal efficacy after respiratory infection of hamsters, which may reflect suboptimal distribution to the lung. Importantly, neither compound induced intestinal toxicity, which was observed as the major adverse effect in clinical trials of brincidofovir, a prodrug of cidofovir which also contains a C-16 modifier. Notably, we found that there was a significant difference in the nephrotoxicity of the two compounds: USC-087 caused significant kidney toxicity while USC-093 did not, at effective doses. These findings will be valuable guidepoints in the future evolution of this new class of potential prodrugs to treat adenovirus infections.


Subject(s)
Adenine/analogs & derivatives , Adenoviridae Infections , Adenovirus Infections, Human , Organophosphonates , Prodrugs , Tyrosine/analogs & derivatives , Cricetinae , Animals , Humans , Adenovirus Infections, Human/drug therapy , Cidofovir/pharmacology , Cidofovir/therapeutic use , Mesocricetus , Antiviral Agents/therapeutic use , Prodrugs/pharmacology , Prodrugs/therapeutic use , Adenoviridae , Virus Replication , Organophosphonates/pharmacology , Organophosphonates/therapeutic use , Adenoviridae Infections/drug therapy , Cytosine/pharmacology , Cytosine/therapeutic use , Amino Acids/pharmacology , Nucleotides/therapeutic use
9.
J Virol ; 84(21): 11470-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739525

ABSTRACT

We previously identified an adenovirus (Ad) protein named U exon protein (UXP) encoded by a leftward-strand (l-strand) transcription unit. Here we identify and characterize the UXP promoter. Primer extension and RNase protection assays mapped the transcription initiation site at 32 nucleotides upstream of the UXP gene initiation codon. A series of viral mutants with mutations at two putative inverted CCAAT (I-CCAAT) boxes and two E2F sites were generated. With mutants lacking the proximal I-CCAAT box, the UXP mRNA level decreased significantly to 30% of the Ad type 5 (Ad5) mRNA level as measured by quantitative reverse transcription-PCR. Decreased UXP was also observed by immunoblotting and immunofluorescence. UXP mRNA and protein levels were similar to those of Ad5 for mutants lacking the distal I-CCAAT box or both putative E2F sites. Ad DNA levels were similar in mutant- and wild-type Ad5-infected cells during the late stage of infection, strongly suggesting that the decreased UXP mRNA and protein from mutants lacking the proximal I-CCAAT box was due to decreased promoter activity. Electrophoretic mobility shift assays (EMSA) indicated that a cellular factor binds specifically to the proximal I-CCAAT box of the UXP promoter. An in vitro luciferase reporter assay demonstrated that basal promoter activity lies between bp -158 and +30 of the transcription initiation site. No E1A-mediated promoter transactivation was observed in 293 cells compared with A549 cells. Thus, we propose that there is a previously unidentified Ad5 promoter that drives expression of the UXP transcription unit. This promoter is embedded within the gene for fiber, and it contains a proximal I-CCAAT box critical for UXP mRNA transcription.


Subject(s)
Adenoviruses, Human/genetics , Genome, Viral/genetics , Promoter Regions, Genetic , Transcription, Genetic , Viral Proteins/genetics , Cell Line , Humans , Transcription Initiation Site , Viral Proteins/analysis
10.
Proc Natl Acad Sci U S A ; 105(20): 7293-7, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18490659

ABSTRACT

Adenoviruses (Ads) cause a wide array of end-organ and disseminated diseases in severely immunosuppressed patients. For example, approximately 20% of pediatric allogeneic hematopoietic stem cell transplant recipients develop disseminated Ad infection, and the disease proves fatal in as many as 50-80% of these patients. Ad infections are a serious problem for solid-organ transplant recipients and AIDS patients as well. Unfortunately, there are no antiviral drugs approved specifically to treat these infections. A suitable animal model that is permissive for Ad replication would help in the discovery process. Here we identify an animal model to study Ad pathogenesis and the efficacy of antiviral compounds. We show that human serotype 5 Ad (Ad5) causes severe systemic disease in immunosuppressed Syrian hamsters that is similar to that seen in immunocompromised patients. We also demonstrate that hexadecyloxypropyl-cidofovir (CMX001) rescues the hamsters from a lethal challenge with Ad5. The antiviral drug provided protection both prophylactically and when given up to 2 days after i.v. exposure to Ad5. CMX001 acts by reducing Ad replication in key target organs. Thus, the immunosuppressed Syrian hamster is a powerful model to evaluate anti-Ad drugs, and its use can facilitate the entry of drugs such as CMX001 into clinical trials.


Subject(s)
Cytosine/analogs & derivatives , Immunosuppressive Agents/therapeutic use , Organophosphonates/pharmacology , Adenoviridae/metabolism , Adenoviridae Infections/metabolism , Adenoviridae Infections/therapy , Animals , Antiviral Agents/pharmacology , Cricetinae , Cytosine/pharmacology , Disease Models, Animal , Drug Screening Assays, Antitumor , Hematopoietic Stem Cells/cytology , Humans , Liver/metabolism , Mesocricetus , Models, Biological
11.
J Virol ; 83(5): 2130-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19073718

ABSTRACT

Immune responses against adenovirus (Ad) vectors pose a possible concern for the outcome of treatment efficacy. To address the role of preexisting immunity in oncolytic Ad vector antitumor efficacy following intratumoral injection of vector as well as tumor-to-tissue spread of the vector, we employed the Syrian hamster model. These animals are immunocompetent, and their tumors and tissues are permissive for replication of Ad type 5 (Ad5). We used the adenovirus death protein-overexpressing Ad5-based vector INGN 007. Subcutaneous tumors were established in groups of hamsters that were or were not immunized with Ad5. Half of the hamsters in these groups were immunosuppressed with cyclophosphamide. For all groups, tumors injected with INGN 007 grew significantly more slowly than those injected with buffer. Under immunocompetent conditions, there was no significant effect of preexisting immunity on vector antitumor efficacy. Soon after the tumors in naïve animals were injected with vector, the hamsters developed neutralizing antibody (NAb) and the difference in NAb titers between the naïve and immunized groups diminished. Under immunosuppressed conditions, preexisting NAb did significantly reduce vector efficacy. Thus, NAb do reduce vector efficacy to some extent, but immunosuppression is required to observe the effect. Regarding vector toxicity, there was spillover of vector from the tumor to the liver and lungs in naïve immunocompetent hamsters, and this was nearly eliminated in the immunized hamsters. Thus, preexisting immunity to Ad5 does not affect INGN 007 antitumor efficacy following intratumoral injection, but immunity prevents vector spillover from the tumor to the liver and lungs.


Subject(s)
Genetic Vectors/therapeutic use , Neoplasms, Experimental/therapy , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antibodies, Viral/immunology , Cell Line, Tumor , Cricetinae , Cyclophosphamide/pharmacology , Humans , Immunocompetence , Immunosuppression Therapy , Liver/virology , Lung/virology , Mesocricetus , Models, Animal , Neoplasms, Experimental/immunology , Neoplasms, Experimental/virology , Virus Replication
12.
Mol Ther ; 17(10): 1724-32, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19602998

ABSTRACT

We have used Syrian hamsters to examine the role of pre-existing immunity to adenovirus (Ad) 5 in the toxicity of the oncolytic Ad vector INGN 007. Groups of hamsters were or were not immunized with Ad5. Half the hamsters were immunosuppressed using cyclophosphamide (CP), then injected intravenously (i.v.) with 3x the maximum tolerated dose (MTD) of INGN 007 (in immunocompetent hamsters), and toxicity and vector replication in the liver were quantitated. In nonimmunized immunocompetent hamsters, toxicity was observed early but the hamsters recovered by day 6 after vector injection. In nonimmunized immunosuppressed hamsters, the vector was lethal by 3 days. Pre-existing neutralizing antibody (NAb) prevented liver infection and hepatotoxicity in both immunocompetent and immunosuppressed hamsters. In another study, passive immunization of immunosuppressed hamsters 1 day before a lethal dose (1x MTD) of INGN 007 prevented liver infection and replication, but immunization 1 day after vector administration was barely effective. When immunosuppressed hamsters were passively immunized 1 day after injection of 1/3rd the MTD of INGN 007, then significant protection was observed against liver infection and toxicity. Therefore, serum NAb are sufficient to prevent oncolytic Ad vector liver infection and toxicity. We saw no evidence that pre-existing immunity was associated with increased vector toxicity.


Subject(s)
Adenoviridae/immunology , Genetic Vectors/immunology , Genetic Vectors/toxicity , Immunity/physiology , Animals , Antibodies, Neutralizing/immunology , Cricetinae , Cyclophosphamide/pharmacology , Female , Immunity/drug effects , Immunosuppressive Agents/pharmacology , Liver/drug effects , Liver/metabolism , Mesocricetus , Oncolytic Virotherapy/adverse effects , Oncolytic Virotherapy/methods
13.
Hum Vaccin Immunother ; 16(3): 636-644, 2020 03 03.
Article in English | MEDLINE | ID: mdl-31584324

ABSTRACT

Treatments with cytotoxic agents or viruses may cause Immunogenic Cell Death (ICD) that immunize tumor-bearing hosts but do not cause complete regression of tumor. We postulate that combining two ICD inducers may cause durable regression in immunocompetent mice. ICD was optimized in vitro by maximizing calreticulin externalization in human colorectal carcinoma (CRC) cells by exposure to mixtures of Oxaliplatin (OX) and human adenovirus (AdV). Six mm diameter CT26 or 4T1 carcinomas in flanks of BALB/c mice were injected once intratumorally (IT) with OX, AdV or their mixture. Tumor growth, Tumor-Infiltrating Lymphocytes (TIL), nodal cytotoxicity, and rejection of a viable cell challenge were measured. Tumors injected IT once with an optimum mixture of 80 µM OX - AdV 25 Multiplicity of Infection (MOI) in PBS buffer were 17-29% the volume of control tumors. When buffer was changed from PBS to 5% dextrose in water (D5W), volumes of tumors injected IT with 80 µM OX-AdV 25 MOI were 10% while IT OX or AdV alone were 32% and 40% the volume of IT buffer-treated tumors. OX-AdV IT increased CD3+ TIL by 4-fold, decreased CD8+ PD-1+ TIL from 79% to 19% and induced cytotoxicity to CT26 cells in draining node lymphocytes while lymphocytes from CT26-bearing untreated mice were not cytotoxic. OX-AdV IT in D5W caused complete regression in 40% of mice. Long-term survivors rejected a contralateral challenge of CT26. The buffer for Oxaliplatin is critical. The two ICD inducer mixture is promising as an agnostic sensitizer for carcinomas like colorectal carcinoma.


Subject(s)
Colorectal Neoplasms , Immunogenic Cell Death , Adenoviridae/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Oxaliplatin
14.
Dis Model Mech ; 13(8)2020 08 27.
Article in English | MEDLINE | ID: mdl-32651192

ABSTRACT

Model animals are indispensable for the study of human diseases, and in general, of complex biological processes. The Syrian hamster is an important model animal for infectious diseases, behavioral science and metabolic science, for which more experimental tools are becoming available. Here, we describe the generation and characterization of an interleukin-2 receptor subunit gamma (Il2rg) knockout (KO) Syrian hamster strain. In humans, mutations in IL2RG can result in a total failure of T and natural killer (NK) lymphocyte development and nonfunctional B lymphocytes (X-linked severe combined immunodeficiency; XSCID). Therefore, we sought to develop a non-murine model to study XSCID and the infectious diseases associated with IL2RG deficiency. We demonstrated that the Il2rg KO hamsters have a lymphoid compartment that is greatly reduced in size and diversity, and is impaired in function. As a result of the defective adaptive immune response, Il2rg KO hamsters developed a more severe human adenovirus infection and cleared virus less efficiently than immune competent wild-type hamsters. Because of this enhanced virus replication, Il2rg KO hamsters developed more severe adenovirus-induced liver pathology than wild-type hamsters. This novel hamster strain will provide researchers with a new tool to investigate human XSCID and its related infections.


Subject(s)
Adaptive Immunity , Adenovirus Infections, Human/virology , Adenoviruses, Human/pathogenicity , Immunocompromised Host , Interleukin Receptor Common gamma Subunit/genetics , X-Linked Combined Immunodeficiency Diseases/genetics , A549 Cells , Adenovirus Infections, Human/genetics , Adenovirus Infections, Human/immunology , Adenovirus Infections, Human/metabolism , Adenoviruses, Human/growth & development , Animals , Animals, Genetically Modified , Disease Models, Animal , Female , Gene Knockout Techniques , HEK293 Cells , Host-Pathogen Interactions , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Liver/immunology , Liver/metabolism , Liver/virology , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/virology , Male , Mesocricetus/genetics , Viral Load , Virus Replication , X-Linked Combined Immunodeficiency Diseases/immunology , X-Linked Combined Immunodeficiency Diseases/metabolism
15.
J Virol ; 82(24): 12510-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18842715

ABSTRACT

A molecular clone of yellow fever virus (YFV) strain 17D was used to identify critical determinants of mouse neuroinvasiveness previously localized to domain III of the neuroadapted SPYF-MN virus envelope protein. Three candidate virulence substitutions (305F-->V, 326K-->E, and 380R-->T) were individually evaluated for their roles in this phenotype in a SCID mouse model. The virus containing a glutamic acid residue at position 326 of the envelope protein (326E) caused rapidly lethal encephalitis, with a mortality rate and average survival time resembling those of the parental SPYF-MN virus. Determinants at positions 380 (380T) and 305 (305V) did not independently affect neuroinvasiveness. Testing a panel of viruses with various amino acid substitutions at position 326 revealed that attenuation of neuroinvasiveness required a positively charged residue (lysine or arginine) at this position. Molecular-modeling studies suggest that residues 326 and 380 contribute to charge clusters on the lateral surface of domain III that constitute putative heparin binding sites, as confirmed by studies of heparin inhibition of plaque formation. The neuroinvasiveness of YFVs in the SCID model correlated inversely with sensitivity to heparin. These findings establish that residue 326 in domain III of the E protein is a critical determinant of YFV neuroinvasiveness in the SCID mouse model. Together with modeling of domain III from virulent YFV strains, the data suggest that heparin binding activity involving lysine at position 326 may be a modulator of YFV virulence phenotypes.


Subject(s)
Adaptation, Biological , Encephalitis/metabolism , Encephalitis/virology , Heparin/metabolism , Viral Structural Proteins/metabolism , Yellow fever virus/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Disease Models, Animal , Encephalitis/genetics , Encephalitis, Japanese , Mice , Mice, SCID , Models, Molecular , Mutation/genetics , Protein Binding , Protein Structure, Tertiary , Structural Homology, Protein , Survival Rate , Viral Structural Proteins/chemistry , Viral Structural Proteins/genetics , Yellow fever virus/genetics , Yellow fever virus/pathogenicity
16.
Mol Ther ; 16(10): 1665-73, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18665155

ABSTRACT

We recently described an immunocompetent Syrian hamster model for oncolytic adenoviruses (Ads) that permits virus replication in tumor cells as well as some normal tissues. This model allows exploration of interactions between the virus, tumor, normal organs, and host immune system that could not be examined in the immunodeficient or nonpermissive animal models previously used in the oncolytic Ad field. Here we asked whether the immune response to oncolytic Ad enhances or limits antitumor efficacy. We first determined that cyclophosphamide (CP) is a potent immunosuppressive agent in the Syrian hamster and that CP alone had no effect on tumor growth. Importantly, we found that the antitumor efficacy of oncolytic Ads was significantly enhanced in immunosuppressed animals. In animals that received virus therapy plus immunosuppression, significant differences were observed in tumor histology, and in many cases little viable tumor remained. Notably, we also determined that immunosuppression allowed intratumoral virus levels to remain elevated for prolonged periods. Although favorable tumor responses can be achieved in immunocompetent animals, the rate of virus clearance from the tumor may lead to varied antitumor efficacy. Immunosuppression, therefore, allows sustained Ad replication and oncolysis, which leads to substantially improved suppression of tumor growth.


Subject(s)
Adenoviridae/genetics , Neoplasms, Experimental/therapy , Oncolytic Virotherapy , Virus Replication/immunology , Adenoviridae/immunology , Adenoviridae/physiology , Animals , Antineoplastic Agents/therapeutic use , Cell Division , Cricetinae , Cyclophosphamide/therapeutic use , Immunohistochemistry , Mesocricetus , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neutralization Tests
17.
FEMS Microbiol Rev ; 43(4): 380-388, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30916746

ABSTRACT

The symptoms of human adenovirus infections are generally mild and self-limiting. However, these infections have been gaining importance in recent years because of a growing number of immunocompromised patients. Solid organ and hematopoietic stem cell transplant patients are subjected to severe immunosuppressive regimes and cannot efficaciously eliminate virus infections. In these patients, adenovirus infections can develop into deadly multi-organ disseminated disease. Presently, in the absence of approved therapies, physicians rely on drugs developed for other purposes to treat adenovirus infections. As there is a need for anti-adenoviral therapies, researchers have been developing new agents and repurposing existing ones to treat adenovirus infections. There are several small molecule drugs that are being tested for their efficacy against human adenoviruses; some of these have reached clinical trials, while others are still in the preclinical phase. Besides these compounds, research on immunotherapy against adenoviral infection has made significant progress, promising another modality for treatment. The availability of an animal model confirmed the activity of some drugs already in clinical use while proving that others are inactive. This led to the identification of several lead compounds that await further development. In the present article, we review the current status of anti-adenoviral therapies and their advancement by in vivo studies in the Syrian hamster model.


Subject(s)
Adenoviridae Infections/drug therapy , Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Development , Mesocricetus , Animals , Cricetinae , Disease Models, Animal
18.
J Virol ; 81(23): 12918-26, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17881437

ABSTRACT

A short open reading frame named the "U exon," located on the adenovirus (Ad) l-strand (for leftward transcription) between the early E3 region and the fiber gene, is conserved in mastadenoviruses. We have observed that Ad5 mutants with large deletions in E3 that infringe on the U exon display a mild growth defect, as well as an aberrant Ad E2 DNA-binding protein (DBP) intranuclear localization pattern and an apparent failure to organize replication centers during late infection. Mutants in which the U exon DNA is reconstructed have a reversed phenotype. Chow et al. (L. T. Chow et al., J. Mol. Biol. 134:265-303, 1979) described mRNAs initiating in the region of the U exon and spliced to downstream sequences in the late DBP mRNA leader and the DBP-coding region. We have cloned this mRNA (as cDNA) from Ad5 late mRNA; the predicted protein is 217 amino acids, initiating in the U exon and continuing in frame in the DBP leader and in the DBP-coding region but in a different reading frame from DBP. Polyclonal and monoclonal antibodies generated against the predicted U exon protein (UXP) showed that UXP is approximately 24K in size by immunoblot and is a late protein. At 18 to 24 h postinfection, UXP is strongly associated with nucleoli and is found throughout the nucleus; later, UXP is associated with the periphery of replication centers, suggesting a function relevant to Ad DNA replication or RNA transcription. UXP is expressed by all four species C Ads. When expressed in transient transfections, UXP complements the aberrant DBP localization pattern of UXP-negative Ad5 mutants. Our data indicate that UXP is a previously unrecognized protein derived from a novel late l-strand transcription unit.


Subject(s)
Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Viral Proteins/genetics , Viral Proteins/physiology , Adenovirus E2 Proteins/analysis , Cell Nucleolus/chemistry , Cell Nucleus/chemistry , Cloning, Molecular , Humans , Immunoblotting , Molecular Sequence Data , Molecular Weight , Open Reading Frames , RNA, Messenger/genetics , RNA, Viral/genetics , Sequence Analysis, DNA , Sequence Deletion , Viral Proteins/biosynthesis , Viral Proteins/chemistry , Virus Replication/physiology
19.
Mol Ther ; 15(3): 598-607, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17191072

ABSTRACT

Novel approaches are needed to improve the antitumor potency and to increase the cancer specificity of oncolytic adenoviruses (Ad). We hypothesized that the combination of interferon-alpha (IFN-alpha) expression with a specific mutation in the e1a gene of Ad could target vector replication to genetic defects in the IFN-alpha pathway resulting in both improved antitumor efficacy and reduced toxicity. The conditionally replicative Ad vector KD3-IFN carries the dl1101/1107 mutation in the e1a gene that eliminates binding of E1A proteins to p300/CBP and pRb. KD3-IFN expresses human IFN-alpha in concurrence with vector replication and overexpresses the adenovirus death protein (ADP; E3-11.6K). The antitumor activity of KD3-IFN was significantly higher than that of a control vector in established human hepatocellular carcinoma tumors in immunodeficient mice and in hamster kidney cancer tumors in immunocompetent Syrian hamsters. The dl1101/1107 mutation rendered Ad replication sensitive to the antiviral effect of IFN-alpha in normal as opposed to cancer cells. These results translated to reduced vector toxicity upon systemic administration to C57BL/6 mice. The combination of Ad oncolysis, ADP overexpression, and IFN-alpha-mediated immunotherapy represents a three-pronged approach for increasing the anticancer efficacy of replicative Ads. Exploiting the dl1101/1107 mutation provides a mechanism for additional selectivity of IFN-alpha-expressing replication-competent Ads.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/metabolism , Interferon-alpha/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Oncolytic Viruses/genetics , Adenovirus E1A Proteins/genetics , Animals , Cell Line , Cell Survival , Cricetinae , Female , Gene Expression , Genetic Therapy , Genetic Vectors/genetics , Genome, Viral/genetics , Interferon-alpha/genetics , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Mice , Mice, Nude , Mutation/genetics , Survival Rate , Time Factors , Transgenes/genetics , Virus Replication , Xenograft Model Antitumor Assays
20.
Mol Ther ; 15(3): 598-607, 2007 Mar.
Article in English | MEDLINE | ID: mdl-28182898

ABSTRACT

Novel approaches are needed to improve the antitumor potency and to increase the cancer specificity of oncolytic adenoviruses (Ad). We hypothesized that the combination of interferon-alpha (IFN-α) expression with a specific mutation in the e1a gene of Ad could target vector replication to genetic defects in the IFN-α pathway resulting in both improved antitumor efficacy and reduced toxicity. The conditionally replicative Ad vector KD3-IFN carries the dl1101/1107 mutation in the e1a gene that eliminates binding of E1A proteins to p300/CBP and pRb. KD3-IFN expresses human IFN-α in concurrence with vector replication and overexpresses the adenovirus death protein (ADP; E3-11.6K). The antitumor activity of KD3-IFN was significantly higher than that of a control vector in established human hepatocellular carcinoma tumors in immunodeficient mice and in hamster kidney cancer tumors in immunocompetent Syrian hamsters. The dl1101/1107 mutation rendered Ad replication sensitive to the antiviral effect of IFN-α in normal as opposed to cancer cells. These results translated to reduced vector toxicity upon systemic administration to C57BL/6 mice. The combination of Ad oncolysis, ADP overexpression, and IFN-α-mediated immunotherapy represents a three-pronged approach for increasing the anticancer efficacy of replicative Ads. Exploiting the dl1101/1107 mutation provides a mechanism for additional selectivity of IFN-α-expressing replication-competent Ads.

SELECTION OF CITATIONS
SEARCH DETAIL