Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Euro Surveill ; 28(19)2023 05.
Article in English | MEDLINE | ID: mdl-37166759

ABSTRACT

IntroductionEmpirical therapy for the treatment of urinary tract infections should be tailored to the current distribution and susceptibility of potential pathogens to ensure optimal treatment.AimWe aimed to provide an up-to-date overview of the epidemiology and susceptibility of Enterobacterales isolated from urine in Germany.MethodsWe retrospectively analysed antimicrobial susceptibility data from 201,152 urine specimens collected between January 2016 and June 2021 from in- and outpatients. Multiple logistic regression analysis was used to evaluate the association between year of investigation and antibiotic resistance, adjusted for age, sex and species subgroup. Subgroup analyses were performed for midstream urine samples obtained from (i) female outpatients aged 15 to 50 years, (ii) female outpatients older than 50 years and (iii) male outpatients.ResultsResistance rates of less than 20% were observed for nitroxoline (3.9%), fosfomycin (4.6%), nitrofurantoin (11.7%), cefuroxime (13.5%) and ciprofloxacin (14.2%). Resistance to trimethoprim/sulfamethoxazole (SXT) (20.1%), amoxicillin-clavulanic acid (20.5%), trimethoprim (24.2%), pivmecillinam (29.9%) and ampicillin (53.7%) was considerably higher. In the subgroup of outpatient women aged 15-50 years, resistance rates were generally lower. Resistance rates of all antibiotics decreased from 2016 to 2021. Multiple logistic regression revealed the lowest adjusted odds ratio (ORadj) of 0.838 (95% confidence interval (CI): 0.819-0.858; p < 0.001) for pivmecillinam and the highest ORadj of 0.989 (95% CI: 0.972-1.007; p = 0.226) for nitrofurantoin.ConclusionsResistance has generally decreased over the past years, independent of sex, age and causative pathogen. Our data provide an important basis for empirical antibiotic recommendations in various settings and patient collectives.


Subject(s)
Amdinocillin Pivoxil , Escherichia coli Infections , Urinary Tract Infections , Female , Male , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Nitrofurantoin/therapeutic use , Amdinocillin Pivoxil/therapeutic use , Retrospective Studies , Escherichia coli , Drug Resistance, Bacterial , Urinary Tract Infections/drug therapy , Urinary Tract Infections/epidemiology , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use , Germany/epidemiology , Microbial Sensitivity Tests , Escherichia coli Infections/drug therapy
2.
Dev Biol ; 486: 26-43, 2022 06.
Article in English | MEDLINE | ID: mdl-35337795

ABSTRACT

The formation of appropriately patterned blood vessel networks requires endothelial cell migration and proliferation. Signaling through the Vascular Endothelial Growth Factor A (VEGFA) pathway is instrumental in coordinating these processes. mRNA splicing generates short (diffusible) and long (extracellular matrix bound) Vegfa isoforms. The differences between these isoforms in controlling cellular functions are not understood. In zebrafish, vegfaa generates short and long isoforms, while vegfab only generates long isoforms. We found that mutations in vegfaa had an impact on endothelial cell (EC) migration and proliferation. Surprisingly, mutations in vegfab more strongly affected EC proliferation in distinct blood vessels, such as intersegmental blood vessels in the zebrafish trunk and central arteries in the head. Analysis of downstream signaling pathways revealed no change in MAPK (ERK) activation, while inhibiting PI3 kinase signaling phenocopied vegfab mutant phenotypes in affected blood vessels. Together, these results suggest that extracellular matrix bound Vegfa might act through PI3K signaling to control EC proliferation in a distinct set of blood vessels during angiogenesis.


Subject(s)
Vascular Endothelial Growth Factor A , Zebrafish , Animals , Cell Proliferation , Neovascularization, Physiologic/genetics , Phenotype , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/genetics , Zebrafish/metabolism
4.
Viruses ; 13(8)2021 07 27.
Article in English | MEDLINE | ID: mdl-34452325

ABSTRACT

In their recent article published in Viruses, Michel Drancourt and colleagues [...].


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Tomography, X-Ray Computed
5.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Article in English | MEDLINE | ID: mdl-33574063

ABSTRACT

To study the mechanisms controlling front-rear polarity in migrating cells, we used zebrafish primordial germ cells (PGCs) as an in vivo model. We find that polarity of bleb-driven migrating cells can be initiated at the cell front, as manifested by actin accumulation at the future leading edge and myosin-dependent retrograde actin flow toward the other side of the cell. In such cases, the definition of the cell front, from which bleb-inhibiting proteins such as Ezrin are depleted, precedes the establishment of the cell rear, where those proteins accumulate. Conversely, following cell division, the accumulation of Ezrin at the cleavage plane is the first sign for cell polarity and this aspect of the cell becomes the cell back. Together, the antagonistic interactions between the cell front and back lead to a robust polarization of the cell. Furthermore, we show that chemokine signaling can bias the establishment of the front-rear axis of the cell, thereby guiding the migrating cells toward sites of higher levels of the attractant. We compare these results to a theoretical model according to which a critical value of actin treadmilling flow can initiate a positive feedback loop that leads to the generation of the front-rear axis and to stable cell polarization. Together, our in vivo findings and the mathematical model, provide an explanation for the observed nonoriented migration of primordial germ cells in the absence of the guidance cue, as well as for the directed migration toward the region where the gonad develops.


Subject(s)
Actins/metabolism , Cell Movement , Cell Polarity , Chemokines/metabolism , Zebrafish Proteins/metabolism , Animals , Cytoskeletal Proteins/metabolism , Germ Cells/cytology , Germ Cells/metabolism , Protein Transport , Zebrafish
6.
Circ Res ; 128(5): 619-635, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33375812

ABSTRACT

RATIONALE: The mechanisms underlying atrial fibrillation (AF), the most common clinical arrhythmia, are poorly understood. Nucleoplasmic Ca2+ regulates gene expression, but the nature and significance of nuclear Ca2+-changes in AF are largely unknown. OBJECTIVE: To elucidate mechanisms by which AF alters atrial-cardiomyocyte nuclear Ca2+ ([Ca2+]Nuc) and CaMKII (Ca2+/calmodulin-dependent protein kinase-II)-related signaling. METHODS AND RESULTS: Atrial cardiomyocytes were isolated from control and AF dogs (kept in AF by atrial tachypacing [600 bpm × 1 week]). [Ca2+]Nuc and cytosolic [Ca2+] ([Ca2+]Cyto) were recorded via confocal microscopy. Diastolic [Ca2+]Nuc was greater than [Ca2+]Cyto under control conditions, while resting [Ca2+]Nuc was similar to [Ca2+]Cyto; both diastolic and resting [Ca2+]Nuc increased with AF. IP3R (Inositol-trisphosphate receptor) stimulation produced larger [Ca2+]Nuc increases in AF versus control cardiomyocytes, and IP3R-blockade suppressed the AF-related [Ca2+]Nuc differences. AF upregulated nuclear protein expression of IP3R1 (IP3R-type 1) and of phosphorylated CaMKII (immunohistochemistry and immunoblot) while decreasing the nuclear/cytosolic expression ratio for HDAC4 (histone deacetylase type-4). Isolated atrial cardiomyocytes tachypaced at 3 Hz for 24 hours mimicked AF-type [Ca2+]Nuc changes and L-type calcium current decreases versus 1-Hz-paced cardiomyocytes; these changes were prevented by IP3R knockdown with short-interfering RNA directed against IP3R1. Nuclear/cytosolic HDAC4 expression ratio was decreased by 3-Hz pacing, while nuclear CaMKII phosphorylation was increased. Either CaMKII-inhibition (by autocamtide-2-related peptide) or IP3R-knockdown prevented the CaMKII-hyperphosphorylation and nuclear-to-cytosolic HDAC4 shift caused by 3-Hz pacing. In human atrial cardiomyocytes from AF patients, nuclear IP3R1-expression was significantly increased, with decreased nuclear/nonnuclear HDAC4 ratio. MicroRNA-26a was predicted to target ITPR1 (confirmed by luciferase assay) and was downregulated in AF atrial cardiomyocytes; microRNA-26a silencing reproduced AF-induced IP3R1 upregulation and nuclear diastolic Ca2+-loading. CONCLUSIONS: AF increases atrial-cardiomyocyte nucleoplasmic [Ca2+] by IP3R1-upregulation involving miR-26a, leading to enhanced IP3R1-CaMKII-HDAC4 signaling and L-type calcium current downregulation. Graphic Abstract: A graphic abstract is available for this article.


Subject(s)
Atrial Fibrillation/metabolism , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Myocytes, Cardiac/metabolism , Action Potentials , Animals , Atrial Fibrillation/physiopathology , Calcium Channels, L-Type/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Dogs , Histone Deacetylases/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/physiology
7.
Phytomedicine ; 17(7): 540-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19962871

ABSTRACT

Screening of a panel of purified compounds isolated from Aglaia sp. (Meliaceae) for inhibition of early steps in the lentiviral replication cycle led to the identification of the 3, 4-secodammarane triterpenoid, ignT1, which inhibited HIV-1 infection potently (IC(50)=0.48microg/ml), while cytotoxic effects and inhibition of cell proliferation were only observed at concentrations exceeding 10.69microg/ml. Time of addition experiments revealed similar kinetics to the non-nucleoside RT-inhibitor (NNRTI), Nevirapine, although the latter was significantly less cytotoxic. However, unlike Nevirapine, dammarenolic acid also potently inhibited the in vitro replication of other retroviruses, including Simian immunodeficiency virus and Murine leukemic virus in vector-based antiviral screening studies. Interestingly, the methyl ester analogue of dammarenolic acid-methyldammarenolate had no anti-HIV-1 activity. Cell cycle analysis revealed that ignT1 arrests HeLa cells at the S and G2/M phase. These results strongly suggest that dammarenolic acid could be a promising lead compound for the development of novel anti-retrovirals.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/drug therapy , HIV-1/drug effects , Phytotherapy , Plant Extracts/therapeutic use , Triterpenes/therapeutic use , Aglaia , Animals , Anti-Retroviral Agents/isolation & purification , Anti-Retroviral Agents/pharmacokinetics , Anti-Retroviral Agents/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cytotoxins , Esters , Haplorhini , HeLa Cells , Humans , Mice , Microbial Sensitivity Tests , Molecular Structure , Nevirapine/pharmacokinetics , Nevirapine/pharmacology , Nevirapine/therapeutic use , Plant Bark , Plant Extracts/adverse effects , Plant Extracts/pharmacology , Retroviridae/drug effects , Simian Immunodeficiency Virus/drug effects , Triterpenes/isolation & purification , Triterpenes/pharmacology
8.
World J Gastroenterol ; 14(12): 1842-50, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18350621

ABSTRACT

AIM: To promote the development of improved tumor vaccination strategies relying on the intratumoral expression of viral fusogenic membrane proteins, we elucidated whether the size of syncytia or the way tumor cells die has an effect on the therapeutic outcome. METHODS: In two syngeneic subcutaneous murine colon cancer models we assessed the anti-neoplastic effect on vector-treated and contralateral untreated tumors. RESULTS: Intratumoral injection of a replication-defective adenovirus encoding respiratory syncytial virus fusion protein (RSV-F) alone (Ad.RSV-F) or together with the attachment glycoprotein RSV-G (Ad.RSV-F/G) led to a significant growth reduction of the vector-treated and contralateral untreated tumors. The treatment response was associated with a strong tumor-specific CTL response and significantly improved survival with medians of 46 d and 44 d, respectively. Intratumoral injection of Ad.RSV-G or a soluble RSV-F encoding adenovirus (Ad.RSV-F(sol)) had no significant anti-neoplastic effect. The median survival of these treatment groups and of Ad.Null-treated control animals was about 30 d. CONCLUSION: Although in vitro transduction of colon cancer cell lines with Ad.RSV-F/G resulted in about 8-fold larger syncytia than with Ad.RSV-F, the in vivo outcome was not significantly different. Transduction of murine colon cancer cell lines with Ad.RSV-F or Ad.RSV-F/G caused apoptotic cell death, in contrast to transduction with Ad.RSV-G or Ad.RSV-F(sol), suggesting an importance of the mode of cell death. Overall, these findings provide insight into improved tumor vaccination strategies relying on the intratumoral expression of viral fusogenic membrane proteins.


Subject(s)
Apoptosis/physiology , Giant Cells , Respiratory Syncytial Viruses/metabolism , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Antineoplastic Agents/metabolism , Cell Fusion , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/metabolism , Giant Cells/cytology , Giant Cells/physiology , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics
9.
Vaccine ; 26(5): 716-26, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-18160188

ABSTRACT

We evaluated the suitability of the Friend Virus (FV) model for the development of improved adenovirus vectors for anti-retroviral vaccination using two types of adenovirus vectors, encoding F-MuLV Env and Gag, which differed only in their fiber genes (Ad5 and Ad5F35). Genetically FV-resistant C57BL/6 mice and highly susceptible CB6F1 hybrid mice were vaccinated by either homologous or heterologous prime-boost regimen. After FV challenge, viral loads in the spleens of C57BL/6 mice were reduced approximately 250-fold and were below the detection threshold in >50% of the mice. Vaccination outcome was critically influenced by the route of vector administration. In CB6F1 mice, vaccination resulted in reduced viremia, delayed onset of splenomegaly, and induction of FV-specific T cells as assessed by tetramer staining. Heterologous prime-boost vaccination resulted in significantly higher neutralizing antibody titers, translating into improved immune protection, in contrast to coexpression of cytokines. Our results suggest that the FV model can provide insight into the development of improved adenovirus vectors for HIV-1 vaccination.


Subject(s)
AIDS Vaccines/administration & dosage , Adenoviridae/genetics , Friend murine leukemia virus/genetics , Friend murine leukemia virus/immunology , Genetic Vectors , Retroviridae Infections/prevention & control , Tumor Virus Infections/prevention & control , Vaccination , AIDS Vaccines/immunology , Adenoviridae/immunology , Animals , Antibodies, Viral/blood , Cytokines/metabolism , Disease Models, Animal , Female , Friend murine leukemia virus/isolation & purification , Genes, env , Genes, gag , Humans , Immunization Schedule , Injections, Intradermal , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Neutralization Tests , Reassortant Viruses , Retroviridae Infections/immunology , Retroviridae Infections/virology , Species Specificity , Spleen/virology , T-Lymphocytes/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
10.
J Invest Dermatol ; 128(4): 988-98, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17960177

ABSTRACT

Advanced melanoma is associated with poor prognosis warranting the development of new therapeutics, such as oncolytic adenoviruses for immunovirotherapy. Since this approach critically depends on efficient transduction of targeted tumor cells, we screened a panel of 22 different adenovirus types for their internalization efficiency in melanoma cells. We demonstrated that the virions of Ad35, Ad38, and Ad3 have significantly higher internalization efficiency in melanoma cells than Ad5, so far the only adenovirus type used in clinical trials for melanoma. Therefore, we developed a conditionally replication-competent Ad5-based vector with the Ad35 fiber shaft and knob domains (Ad5/35) and compared its therapeutic efficacy with the homologous vector carrying the native Ad5 fiber. To further enhance virotherapy, we combined the oncolytic adenovirus vectors with intratumoral expression of measles virus fusogenic membrane glycoproteins H and F (MV-H/F) and dacarbazine chemotherapy. In a human melanoma xenograft model, established from a short-term culture of primary melanoma cells, we demonstrated that the Ad5/35-based therapy had a significantly greater anti-neoplastic effect than the homologous Ad5-based therapy. Furthermore, the combination of virotherapy, intratumoral expression of MV-H/F, and chemotherapy was clearly superior to single- or double-agent therapy. In conclusion, Ad35-based vectors are promising for the treatment of melanoma.


Subject(s)
Adenoviruses, Human , Genetic Vectors , Melanoma/therapy , Oncolytic Virotherapy , Skin Neoplasms/therapy , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Combined Modality Therapy , Dacarbazine/therapeutic use , Genetic Vectors/genetics , Humans , Melanoma/drug therapy , Membrane Cofactor Protein/analysis , Mice , Mice, Nude , Recoverin/analysis , Skin Neoplasms/drug therapy , Viral Fusion Proteins/genetics , Viral Proteins/genetics , Virus Internalization , Virus Replication , Xenograft Model Antitumor Assays
11.
Int J Mol Med ; 20(5): 673-81, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17912460

ABSTRACT

We assessed whether intratumoral expression of the fusogenic membrane protein of vesicular stomatitis virus (VSV-G), encoded by a replication-defective adenovirus vector (Ad.VSV-G), alone or in combination with local coexpression of cytokines induces tumor-specific immune responses in a syngeneic murine colon cancer model. We confirmed in vitro by dye colocalization that transduction of murine cells with Ad.VSV-G induces cell-cell fusion. In a bilateral syngeneic subcutaneous colon cancer model in C57BL/6 and BALB/c mice, we demonstrated that intratumoral injection of Ad.VSV-G leads to a significant growth reduction of the directly vector-treated tumor, but also of the contralateral not directly vector-treated tumor. When compared to monotherapy, the anti-neoplastic efficacy was significantly enhanced when intratumoral Ad.VSV-G administration was combined with adenovirus vectors encoding IL-2, IL-12, IL-18, IL-21, or GM-CSF. The anti-tumor effects of the first three cytokines in combination with VSV-G expression were somewhat greater than those of the latter two. However, the differences did not reach statistical significance. The combination therapy resulted also in a significantly enhanced survival when compared to monotherapy. In addition, we demonstrated that intratumoral expression of VSV-G in combination with the tested cytokines induced a strong tumor-specific cytotoxic T lymphocyte (CTL) response and infiltration of tumors with macrophages. The effects of the combination therapy were clearly greater than those of the monotherapy. Our experimental data indicate that intratumoral expression of VSV-G, particularly in combination with cytokines, is a promising novel tool for the development of in situ tumor vaccination approaches.


Subject(s)
Adenoviridae/metabolism , Cytokines/metabolism , Genetic Vectors/metabolism , Immunotherapy , Membrane Glycoproteins/metabolism , Neoplasms/immunology , Neoplasms/therapy , Viral Envelope Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Cancer Vaccines , Cell Fusion , Cell Line, Tumor , Cell Movement/drug effects , Colon/drug effects , Colon/pathology , Female , Giant Cells/drug effects , Giant Cells/pathology , Humans , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Splenomegaly/pathology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Treatment Outcome , Xenograft Model Antitumor Assays
12.
Mol Cancer Ther ; 6(7): 1942-50, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17620425

ABSTRACT

Although cancers can naturally elicit immune responses, immune ignorance is a common observation preventing immune-mediated elimination of tumor cells. We assessed whether intratumoral expression of respiratory syncytial virus fusion (RSV-F) protein, encoded by a replication-defective adenovirus vector (Ad.RSV-F), alone or in combination with local coexpression of cytokines can induce tumor-specific immune responses in a syngeneic murine colon cancer model. We confirmed in vitro by dye colocalization that transduction of murine cells with Ad.RSV-F induces cell-cell fusion. In vivo, we showed in a bilateral syngeneic s.c. colon cancer model in C57BL/6 and BALB/c mice that intratumoral injection of Ad.RSV-F leads to a significant volume reduction not only of the directly vector-treated tumor but also of the contralateral not directly vector-treated tumor. The intratumoral administration of Ad.RSV-F in combination with adenovirus vectors encoding interleukin (IL)-2, IL-12, IL-18, IL-21, or granulocyte macrophage colony-stimulating factor significantly enhanced the antitumor effect on the directly vector-treated tumor and also on the contralateral tumor. The antineoplastic efficacy of this combined treatment was significantly higher than that of the individual treatment components and was associated with the induction of a tumor-specific CTL response and increased infiltration of the tumors by natural killer cells and macrophages. Intratumoral coexpression of RSV-F and IL-21 resulted in the highest tumor growth inhibition and improved survival. Our experimental data indicate that intratumoral expression of RSV-F in combination with cytokines is a promising novel tool for the development of in situ tumor vaccination approaches.


Subject(s)
Adenoviridae/genetics , Cancer Vaccines/immunology , Colorectal Neoplasms/prevention & control , Cytokines/genetics , Respiratory Syncytial Viruses , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Animals , Antineoplastic Agents/pharmacology , Cancer Vaccines/genetics , Cell Fusion , Colorectal Neoplasms/immunology , Cytokines/immunology , Female , Genetic Vectors/genetics , HT29 Cells , Humans , Killer Cells, Natural/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Splenomegaly/pathology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects
13.
J Gene Med ; 9(9): 764-78, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17640083

ABSTRACT

Adenovirus type 5 (Ad5)-based vectors have been used in clinical trials for glioblastoma treatment, but the capacity of Ad5 to infect human glioma cells was questioned. Seeking to improve the adenovirus transduction, we tested four Ad5-based vectors differing only in their fiber gene on permanent and short-term cultures of glioblastoma cells. A wild-type fiber Ad5 vector (Ad5.Luc) was compared to an RGD integrin-binding motif-containing fiber adenovirus (AdlucRGD) and the two fiber chimeras Ad5/3 and Ad5/35, with vector binding redirected to the Ad3 or Ad35 receptor, respectively. Compared to Ad5, the transduction of the tested short-term glioblastoma cultures with the vector Ad5/35.Luc, AdlucRGD and Ad5/3.Luc was enhanced by approximately 72%, approximately 13% and approximately 2%, respectively. To limit adenovirus spread, we aimed to develop conditionally replicative Ad5/35 vectors by targeting the expression of the essential E1 and E4 genes; in addition, some vectors had the E1Delta24 deletion. We analyzed eleven promoters for their activity in glioblastoma cells and determined the specificity of eight replicative adenovirus vectors in vitro. We evaluated the most promising vectors with E1/E4 under the control of the GFAP/Ki67 or E2F-1/COX-2 promoters, and the native Ad5 or the chimeric Ad5/35 fiber for their antineoplastic activity in a subcutaneous and intracranial glioblastoma xenograft model. Animals treated with the Ad5/35-based vectors showed significantly smaller tumors and longer survival than those treated with the homologous Ad5 vectors; no significant toxicity was observed in the intracranial model. Our data suggest that Ad5/35-based vectors are promising tools for glioblastoma treatment.


Subject(s)
Adenoviridae/physiology , Genetic Therapy , Glioblastoma/genetics , Glioblastoma/therapy , Viral Proteins/metabolism , Virus Replication , Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , Adenovirus E4 Proteins/genetics , Animals , Cell Line, Tumor , Cytopathogenic Effect, Viral , Female , Humans , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Oncolytic Viruses/metabolism , Organ Specificity , Promoter Regions, Genetic/genetics , Transcription, Genetic , Transduction, Genetic , Xenograft Model Antitumor Assays
14.
World J Gastroenterol ; 13(22): 3063-70, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17589921

ABSTRACT

AIM: To evaluate whether intratumoral expression of measles virus fusogenic membrane glycoproteins H and F (MV-FMG), encoded by an adenovirus vector Ad.MV-H/F, alone or in combination with local coexpression of cytokines (IL-2, IL-12, IL-18, IL-21 or GM-CSF), can serve as a platform for inducing tumor-specific immune responses in colon cancer. METHODS: We used confocal laser scanning microscopy and flow cytometry to analyze cell-cell fusion after expression of MV-FMG by dye colocalization. In a syngeneic bilateral subcutaneous MC38 and Colon26 colon cancer model in C57BL/6 and BALB/c mice, we assessed the effect on both the directly vector-treated tumor as well as the contralateral, not directly vector-treated tumor. We assessed the induction of a tumor-specific cytotoxic T lymphocyte (CTL) response with a lactate dehydrogenase (LDH) release assay. RESULTS: We demonstrated in vitro that transduction of MC38 and Colon26 cells with Ad.MV-H/F resulted in dye colocalization, indicative of cell-cell fusion. In addition, in the syngeneic bilateral tumor model we demonstrated a significant regression of the directly vector-inoculated tumor upon intratumoral expression of MV-FMG alone or in combination with the tested cytokines. We observed the highest anti-neoplastic efficacy with MV-FMG and IL-21 coexpression. The degree of tumor regression of the not directly vector-treated tumor correlated with the anti-neoplastic response of the directly vector-treated tumor. This regression was mediated by a tumor-specific CTL response. CONCLUSION: Our data indicate that intratumoral expression of measles virus fusogenic membrane glycoproteins is a promising tool both for direct tumor treatment as well as for tumor vaccination approaches that can be further enhanced by cytokine coexpression.


Subject(s)
Adenocarcinoma/prevention & control , Adenoviridae/genetics , Cancer Vaccines/therapeutic use , Colonic Neoplasms/prevention & control , Cytokines/metabolism , Genetic Vectors/therapeutic use , Viral Fusion Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Fusion , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytokines/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , HT29 Cells , Humans , Immunotherapy/methods , Interleukins/genetics , Interleukins/metabolism , Macrophages/pathology , Measles virus/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Viral Fusion Proteins/genetics
15.
Hum Gene Ther ; 18(5): 435-50, 2007 May.
Article in English | MEDLINE | ID: mdl-17518612

ABSTRACT

We evaluated whether the expression of measles virus fusogenic membrane glycoproteins H and F (MV-FMG), encoded by a herpes simplex virus type 1 (HSV-1) amplicon vector, can serve with or without viral oncolysis (G47Delta) and facultative irinotecan chemotherapy, alone or in combination with the monoclonal epidermal growth factor receptor (EGFR) inhibitory antibody cetuximab, as a platform for inducing tumor-specific immune responses against colon cancer. We demonstrated in vitro that MV-FMG expression in murine cells resulted in cell-cell fusion and synergistically enhanced the cytotoxicity of irinotecan alone or in combination with cetuximab. In a bilateral syngeneic subcutaneous MC38 and Colon26 tumor model in C57BL/6 and BALB/c mice we assessed both the effect on directly vector-treated tumors and the effect on contralateral, not directly vector-treated tumors. We demonstrated that the combination of three treatment components with or without cetuximab resulted in the best volume reduction of both directly vector-treated and not directly vector-treated tumors as well as pronounced infiltration of both tumor types with natural killer cells, macrophages, and T cells. T cells of these animals exhibited strong ex vivo cytotoxic activity against the tumor cells, indicating that the antineoplastic effect on untreated tumors was mediated by an antitumor immune response. Preexisting immunity against HSV-1 or measles virus had no detrimental effect on overall treatment efficacy. Our data indicate that MV-FMG expression in combination with viral oncolysis with or without clinically relevant chemotherapy for colon cancer treatment warrants further investigation.


Subject(s)
Colonic Neoplasms/therapy , Measles virus/genetics , Oncolytic Virotherapy/methods , Viral Fusion Proteins/genetics , Animals , Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Cell Fusion , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Combined Modality Therapy , Genetic Therapy/methods , Humans , Irinotecan , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology
16.
Cancer Biol Ther ; 6(4): 510-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17374986

ABSTRACT

Previously we demonstrated that the expression of fusogenic membrane proteins (FMG) of measles virus (MV-H/F) can synergistically enhance chemotherapy. In this study, we used median-effect analysis to evaluate whether the expression of respiratory syncytial virus (RSV-F), as well as vesicular stomatitis virus (VSV-G) can also synergistically enhance chemotherapy. Furthermore we elucidated by western blot analysis some molecular pathways that might be responsible for this effect. We showed in colorectal cancer cell lines that the expression of MV-H/F, but also of RSV-F, as well as VSV-G can synergistically enhance p53-independent clinically relevant chemotherapy (FOLFOX) over most of the cytotoxic dose range. In a subcutaneous HT-29 colorectal xenograft model, we demonstrated that the administration of replication-deficient adenovirus vectors encoding MV-H/F, RSV-F or VSV-G in combination with FOLFOX significantly enhanced treatment outcome when compared to the treatment with each compound individually. The anti-neoplastic efficacy of RSV-F was somewhat better than that of MV-H/F and both were statistically significantly more efficacious than VSV-G alone or in combination with chemotherapy. Treatment efficacy was further significantly improved when the replication-deficient FMG encoding vectors were trans-complemented for replication with a replication-restricted oncolytic adenovirus to improve tumor transduction efficiency. The combination of FMG expression, chemotherapy and trans-complementing oncolytic vectors resulted in a significantly better treatment efficacy than treatment with its components as single- or double-agent therapy. Our data indicates that FMG expression (i.e., RSV-F and MV-H/F) in combination with chemotherapy and viral oncolysis warrants further investigations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/therapy , Genetic Therapy , Respiratory Syncytial Viruses/genetics , Vesiculovirus/genetics , Viral Fusion Proteins/genetics , Animals , Annexin A5/metabolism , Apoptosis , Caspases/metabolism , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Combined Modality Therapy , Female , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Membrane Fusion , Mice , Mice, Nude , Mitochondria/enzymology , Organoplatinum Compounds/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
17.
Hum Gene Ther ; 18(1): 51-62, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17184155

ABSTRACT

The clinical course of sarcoma warrants the development of new therapeutic options, such as gene therapy. However, the lack of coxsackievirus-adenovirus receptor (CAR) on sarcoma cells limits the efficacy of adenovirus type 5 (Ad5)-based gene therapy. In this study we evaluated 20 different adenoviral types and 1 Ad5 vector with RGD-containing fiber for their internalization efficiency in sarcoma cells. We demonstrated that adenovirus types 35, 3, 7, 11, 9, and 22 and Ad5lucRGD virions (ranked in descending order) have significantly higher internalization efficiency in the tested sarcoma cells when compared with Ad5. On the basis of these results we developed a conditionally replication-competent adenoviral vector, Ad5Delta24.Ki.COX, and compared its oncolytic efficacy with that of Ad5/35Delta24.Ki.COX, an Ad5-based vector with the Ad35 fiber shaft and knob domains. Because both vectors differed only in the fiber, we were able to assess whether the adenoviral type with the most efficient internalization resulted also in enhanced treatment efficacy. We evaluated the antineoplastic activity of the oncolytic adenoviral vectors alone or in combination with the expression of measles virus fusogenic membrane glycoproteins and/or ifosfamide. The findings of our xenograft model were as follows: animals that received Ad5/35-based therapy had significantly smaller tumors than animals treated with the homologous Ad5-based vectors. In addition, we demonstrated that the combination of virotherapy, intratumoral expression of fusogenic membrane glycoproteins, and ifosfamide was clearly superior compared with treatment with individual components alone or as combinations of two components. In conclusion, Ad35-based vectors are promising for the treatment of sarcoma.


Subject(s)
Adenoviridae , Genetic Therapy , Genetic Vectors , Sarcoma/therapy , Transduction, Genetic , Virus Internalization , Adenoviridae/genetics , Animals , Cell Line, Tumor , Female , Humans , Measles virus/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Oligopeptides/genetics , Sarcoma/genetics , Transplantation, Heterologous , Virus Replication/genetics
18.
BMC Biotechnol ; 6: 36, 2006 Aug 03.
Article in English | MEDLINE | ID: mdl-16887042

ABSTRACT

BACKGROUND: Oncolytic adenoviruses are promising agents for the multimodal treatment of cancer. However, tumor-selectivity is crucial for their applicability in patients. Recent studies by several groups demonstrated that oncolytic adenoviruses with tumor-/tissue-specific expression of the E1 and E4 genes, which are pivotal for adenoviral replication, have a specificity profile that is superior to viruses that solely target the expression of E1 or E4 genes. Presently the E1 and E4 regions are modified in a time consuming sequential fashion. RESULTS: Based on the widely used adenoviral cloning system AdEasy we generated a novel transfer vector that allows efficient and rapid generation of conditionally replication-competent adenovirus type 5 based vectors with the viral E1 and E4 genes under the transcriptional control of heterologous promoters. For insertion of the promoters of interest our transfer vector has two unique multiple cloning sites. Additionally, our shuttle plasmid allows encoding of a transgene within the E1A transcription unit. The modifications, including E1 mutations, are introduced into the adenoviral genome by a single homologous recombination step in Escherichia coli. Subsequently infectious viruses are rescued from plasmids. As a proof-of-concept we generated two conditionally replication-competent adenoviruses Ad.Ki x COX and Ad.COX x Ki with the promoters of the Ki-67 protein and the cyclooxygenase-2 (COX-2) driving E1 and E4 and vice versa. CONCLUSION: We demonstrated with our cloning system efficient generation of double heterologous promoter controlled oncolytic adenoviral vectors by a single homologous recombination step in bacteria. The generated viruses showed preferential replication in tumor cells and in a subcutaneous HT-29 colon cancer xenograft model the viruses demonstrated significant oncolytic activity comparable with dl327.


Subject(s)
Adenoviruses, Human/genetics , Escherichia coli/genetics , Genetic Engineering/methods , Genetic Vectors/genetics , Oncolytic Viruses/genetics , Promoter Regions, Genetic/genetics , Recombination, Genetic/genetics , Adenovirus E1 Proteins/genetics , Adenovirus E1 Proteins/metabolism , Adenovirus E4 Proteins/genetics , Adenovirus E4 Proteins/metabolism , Adenoviruses, Human/physiology , Animals , Child , Cyclooxygenase 2/genetics , Cytopathogenic Effect, Viral , DNA, Recombinant/genetics , HT29 Cells , Humans , Ki-67 Antigen/genetics , Mice , Mice, Inbred BALB C , Mutation/genetics , Oncolytic Viruses/physiology , Organ Specificity , Virus Replication , Xenograft Model Antitumor Assays
19.
Mol Cancer Ther ; 5(8): 2013-22, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16928822

ABSTRACT

Pancreatic cancer has a poor prognosis with an annual mortality rate close to the annual incidence rate. We evaluated whether the expression of measles virus fusogenic membrane glycoproteins (FMG) H and F will enhance chemotherapy. Using Chou-Talalay analysis, we showed in vitro in pancreatic cancer cells that the expression of FMG often synergistically enhances clinically relevant chemotherapy. Furthermore, cell fusion in combination with chemotherapy resulted in strongly enhanced Annexin V binding, an early marker for apoptosis, when compared with single treatment. We showed in an i.p. and s.c. pancreatic xenograft model that the administration of a replication-defective adenoviral vector Ad.H/F encoding tumor-restricted FMG in combination with gemcitabine significantly enhanced treatment outcome when compared with treatment with each compound individually. To improve tumor transduction efficiency, the Ad.H/F vector was also transcomplemented with an oncolytic replication-restricted adenovirus (Ad.COX*MK), resulting in significantly improved treatment efficacy. We assessed treatment efficacy by survival analysis or measuring growth, respectively. In the i.p. model, on day 120, three of eight animals treated with this novel triple therapy consisting of Ad.H/F, gemcitabine, and Ad.COX*MK were alive and tumor free. Treatment with Ad.H/F and Ad.COX*MK resulted in one long-term survivor. In all other treatment groups, there were no long-term survivors. The significantly improved therapeutic outcome of animals receiving the triple therapy was attributed to multiple factors, including most likely improved FMG expression throughout the tumor and enhanced sensitivity of the tumor cells to gemcitabine by adenoviral gene products but also FMG expression. Qualitatively similar results were obtained in a s.c. pancreatic xenograft model.


Subject(s)
Antineoplastic Agents/pharmacology , Membrane Fusion Proteins/genetics , Membrane Glycoproteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Adenoviridae/genetics , Animals , Apoptosis , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Female , Genetic Vectors , Humans , Membrane Fusion Proteins/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/mortality , Survival Rate , Transduction, Genetic , Tumor Cells, Cultured , Gemcitabine
20.
J Virol ; 80(7): 3549-58, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16537623

ABSTRACT

Oncolytic adenoviruses have emerged as a promising approach for the treatment of tumors resistant to other treatment modalities. However, preclinical safety studies are hampered by the lack of a permissive nonhuman host. Screening of a panel of primary cell cultures from seven different animal species revealed that porcine cells support productive replication of human adenovirus type 5 (Ad5) nearly as efficiently as human A549 cells, while release of infectious virus by cells from other animal species tested was diminished by several orders of magnitude. Restriction of productive Ad5 replication in rodent and rabbit cells seems to act primarily at a postentry step. Replication efficiency of adenoviral vectors harboring different E1 deletions or mutations in porcine cells was similar to that in A549 cells. Side-by-side comparison of the viral load kinetics in blood of swine and mice injected with Ad5 or a replication-deficient adenoviral vector failed to provide clear evidence for virus replication in mice. In contrast, evidence suggests that adenovirus replication occurs in swine, since adenoviral late gene expression produced a 13.5-fold increase in viral load in an individual swine from day 3 to day 7 and 100-fold increase in viral DNA levels in the Ad5-infected swine compared to the animal receiving a replication-deficient adenovirus. Lung histology of Ad5-infected swine revealed a severe interstitial pneumonia. Although the results in swine are based on a small number of animals and need to be confirmed, our data strongly suggest that infection of swine with human adenovirus or oncolytic adenoviral vectors is a more appropriate animal model to study adenoviral pathogenicity or pharmacodynamic and toxicity profiles of adenoviral vectors than infection of mice.


Subject(s)
Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , DNA, Viral/administration & dosage , Virus Replication , Adenoviruses, Human/pathogenicity , Animals , Cell Line , Cricetinae , Cytopathogenic Effect, Viral , DNA, Viral/analysis , Female , Flow Cytometry , Genetic Vectors , Green Fluorescent Proteins/metabolism , Guinea Pigs , HeLa Cells , Humans , Inflammation/pathology , Inflammation/virology , Injections, Intravenous , Kidney/cytology , Kidney/virology , Liver/metabolism , Liver/pathology , Liver/virology , Lung/pathology , Lung/virology , Marmota , Mice , Mice, Inbred BALB C , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Sigmodontinae , Sus scrofa , Transduction, Genetic , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...