Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Gene Ther ; 2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38678160

RESUMO

Pseudomonas aeruginosa poses a significant threat to immunocompromised individuals and those with cystic fibrosis. Treatment relies on antibiotics, but persistent infections occur due to intrinsic and acquired resistance of P. aeruginosa towards multiple classes of antibiotics. To date, there are no licensed vaccines for this pathogen, prompting the urgent need for novel treatment approaches to combat P. aeruginosa infection and persistence. Here we validated AAV vectored immunoprophylaxis as a strategy to generate long-term plasma and mucosal expression of highly protective monoclonal antibodies (mAbs) targeting the exopolysaccharide Psl (Cam-003) and the PcrV (V2L2MD) component of the type-III secretion system injectosome either as single mAbs or together as a bispecific mAb (MEDI3902) in a mouse model. When administered intramuscularly, AAV-αPcrV, AAV-αPsl, and AAV-MEDI3902 significantly protected mice challenged intranasally with a lethal dose of P. aeruginosa strains PAO1 and PA14 and reduced bacterial burden and dissemination to other organs. While all AAV-mAbs provided protection, AAV-αPcrV and AAV-MEDI3902 provided 100% and 87.5% protection from a lethal challenge with 4.47 × 107 CFU PAO1 and 87.5% and 75% protection from a lethal challenge with 3 × 107 CFU PA14, respectively. Serum concentrations of MEDI3902 were ~10× lower than that of αPcrV, but mice treated with this vector showed a greater reduction in bacterial dissemination to the liver, lung, spleen, and blood compared to other AAV-mAbs. These results support further investigation into the use of AAV vectored immunoprophylaxis to prevent and treat P. aeruginosa infections and other bacterial pathogens of public health concern for which current treatment strategies are limited.

2.
J Infect Dis ; 228(Suppl 7): S682-S690, 2023 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-37638865

RESUMO

Although there are no approved countermeasures available to prevent or treat disease caused by Marburg virus (MARV), potently neutralizing monoclonal antibodies (mAbs) derived from B cells of human survivors have been identified. One such mAb, MR191, has been shown to provide complete protection against MARV in nonhuman primates. We previously demonstrated that prophylactic administration of an adeno-associated virus (AAV) expressing MR191 protected mice from MARV. Here, we modified the AAV-MR191 coding sequence to enhance efficacy and reevaluated protection in a guinea pig model. Remarkably, 4 different variants of AAV-MR191 provided complete protection against MARV, despite administration 90 days prior to challenge. Based on superior expression kinetics, AAV-MR191-io2, was selected for evaluation in a dose-reduction experiment. The highest dose provided 100% protection, while a lower dose provided ∼88% protection. These data confirm the efficacy of AAV-mediated expression of MR191 and support the further development of this promising MARV countermeasure.


Assuntos
Doença do Vírus de Marburg , Marburgvirus , Humanos , Cobaias , Animais , Camundongos , Linfócitos B , Anticorpos Neutralizantes
3.
Gene Ther ; 30(5): 455-462, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-33608675

RESUMO

Clostridium difficile is the leading cause of antibiotic-associated nosocomial diarrhea in the developed world. When the host-associated colon microbiome is disrupted by the ingestion of antibiotics, C. difficile spores can germinate, resulting in infection. C. difficile secretes enterotoxin A (TcdA) and cytotoxin B (TcdB) that are responsible for disease pathology. Treatment options are limited as the bacterium demonstrates resistance to many antibiotics, and even with antibacterial therapies, recurrences of C. difficile are common. Actotoxumab and bezlotoxumab are human monoclonal antibodies that bind and neutralize TcdA and TcdB, respectively. In 2016, the US food and drug administration (FDA) approved bezlotoxumab for use in the prevention of C. difficile infection recurrence. To ensure the long-term expression of antibodies, gene therapy can be used. Here, adeno-associated virus (AAV)6.2FF, a novel triple mutant of AAV6, was engineered to express either actotoxumab or bezlotoxumab in mice and hamsters. Both antibodies expressed at greater than 90 µg/mL in the serum and were detected at mucosal surfaces in both models. Hundred percent of mice given AAV6.2FF-actoxumab survived a lethal dose of TcdA. This proof of concept study demonstrates that AAV-mediated expression of C. difficile toxin antibodies is a viable approach for the prevention of recurrent C. difficile infections.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Infecções por Clostridium , Humanos , Animais , Camundongos , Toxinas Bacterianas/genética , Anticorpos Neutralizantes , Infecções por Clostridium/prevenção & controle , Infecções por Clostridium/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Antibacterianos/uso terapêutico , Proteínas de Bactérias/genética , Proteínas de Bactérias/uso terapêutico
4.
Gene Ther ; 2023 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-36732618

RESUMO

Respiratory syncytial virus (RSV) causes acute lower respiratory tract infections, with potential lower respiratory tract infections, which can be particularly problematic in infants and the elderly. There are no approved vaccines for RSV. The current standard of care for high-risk individuals is monthly administration of palivizumab, a humanized murine monoclonal antibody (mAb) targeting the RSV fusion protein. Adeno-associated virus (AAV)-mediated expression of mAbs has previously led to sustained expression of therapeutic concentrations of mAbs in several animal models, representing an alternative to repetitive passive administration. Intramuscular (IM) administration of AAV6.2FF expressing RSV antibodies, palivizumab or hRSV90, resulted in high concentrations of human (h)IgG1 mAbs in the serum and at various mucosal surfaces, while intranasal administration limited hIgG expression to the respiratory tract. IM administration of AAV6.2FF-hRSV90 or AAV6.2FF-palivizumab in a murine model provided sterilizing immunity against challenge with RSV A2. Evidence of maternal passive transfer of vectorized hRSV90 was detected in both murine and ovine models, with circulating mAbs providing sterilizing immunity in mouse progeny. Finally, addition of a "kill switch" comprised of LoxP sites flanking the mAb genes resulted in diminished serum hIgG after AAV-DJ-mediated delivery of Cre recombinase to the same muscle group that was originally transduced with the AAV-mAb vector. The ability of this AAV-mAb system to mediate robust, sustained mAb expression for maternal transfer to progeny in murine and ovine models emphasizes the potential of this platform for use as an alternative prophylactic vaccine for protection against neonatal infections, particularly in high-risk infants.

5.
Gene Ther ; 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-36050451

RESUMO

Vectored monoclonal antibody (mAb) expression mediated by adeno-associated virus (AAV) gene delivery leads to sustained therapeutic mAb expression and protection against a wide range of infectious diseases in both small and large animal models, including nonhuman primates. Using our rationally engineered AAV6 triple mutant capsid, termed AAV6.2FF, we demonstrate rapid and robust expression of two potent human antibodies against Marburg virus, MR78 and MR191, following intramuscular (IM) administration. IM injection of mice with 1 × 1011 vector genomes (vg) of AAV6.2FF-MR78 and AAV6.2FF-MR191 resulted in serum concentrations of approximately 141 µg/mL and 195 µg/mL of human IgG, respectively, within the first four weeks. Mice receiving 1 × 1011 vg (high) and 1 × 1010 vg (medium) doses of AAV6.2FF-MR191 were completely protected against lethal Marburg virus challenge. No sex-based differences in serum human IgG concentrations were observed; however, administering the AAV-mAb over multiple injection sites significantly increased serum human IgG concentrations. IM administration of three two-week-old lambs with 5 × 1012 vg/kg of AAV6.2FF-MR191 resulted in serum human IgG expression that was sustained for more than 460 days, concomitant with low levels of anti-capsid and anti-drug antibodies. AAV-mAb expression is a viable method for prolonging the therapeutic effect of recombinant mAbs and represents a potential alternative "vaccine" strategy for those with compromised immune systems or in possible outbreak response scenarios.

6.
Invest New Drugs ; 33(3): 761-74, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25693885

RESUMO

Pelareorep (REOLYSIN®) is an investigational new drug, a proprietary formulation consisting of a live, replication-competent, naturally occurring Reovirus Type 3 Dearing strain. Through several preclinical studies it was determined that reovirus can exhibit profound cytotoxic effects on cancer cells predominantly with an activated RAS-signalling pathway. Moreover, it was discovered that reoviruses can "hitchhike" on peripheral blood mononuclear cells and dendritic cells, thereby evading neutralizing antibodies of the host immune system. Cell carriage, targeted delivery, triggering host immune response and other inherent characteristics of the reovirus led to its further advancement into cancer therapy. When injected into Sprague-Dawley rats, the viral routes of clearance, predominantly through the spleen and liver, remained consistent with earlier studies. Toxicology findings were considered incidental and not associated with pelareorep when tested in animal models. Pelareorep demonstrated a high level of homogeneity at the amino acid level and genetic stability when compared to the master and working virus banks. The drug is manufactured in a 100 L bioreactor after which it is purified and formulated for use in pre-clinical, clinical and research studies. Over the past few decades, we have witnessed a paradigm shift from conventional therapy to the conceivable use of oncolytic viruses for the treatment of cancer. This review will detail pre-clinical evidence of anticancer activity of pelareorep that has led to extensive clinical development. Several Phase I-II clinical trials have been completed or are ongoing in cancer patients on a broad spectrum of solid tumors and hematologic malignancies.


Assuntos
Antineoplásicos/metabolismo , Vírus Oncolíticos/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Ensaios Clínicos como Assunto , Humanos , Imunoterapia , Reoviridae/metabolismo
7.
Appl Microbiol Biotechnol ; 98(4): 1763-70, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24419798

RESUMO

REOLYSIN (pelareorep) is a proprietary isolate of the reovirus T3D (Type 3 Dearing) strain which is currently being tested in clinical trials as an anticancer therapeutic agent. Reovirus genomes are composed of ten segments of double-stranded ribonucleic acid (RNA) characterized by genome size: large (L1, L2, and L3), medium (M1, M2, and M3), and small (S1, S2, S3, and S4). The objective of this work was to evaluate the homogeneity and genetic stability of REOLYSIN. Sanger sequencing (SS) performed on test articles derived from the Master Virus Bank (MVB) and Working Virus Bank (WVB) identified many modifications when compared to GenBank reference sequences. Massively parallel sequencing (MPS) using Roche-454 sequencing was performed on REOLYSIN (100 L scale) and resulted in 69,821,115 bases and an average of 335 bases per read. Twenty-nine high confidence differences relative to the GenBank reference sequence were identified in REOLYSIN by MPS. Of those, 27 were previously identified by SS in the virus bank-derived test articles. Of the remaining two nucleotide differences, one was predicted to be silent at the amino acid level (L3 genome-T3163C, codon 1054, 86% of the population was "T" and 13% of the population were reported as "C"). The other modification was in the noncoding region (M1 genome-A2284A to A2284G), and A2284G was present in 97% of the population. The results obtained from MPS were comparable to those from SS; both demonstrate a high level of homogeneity at the amino acid level and genetic stability of REOLYSIN. Finally, phylogenetic analysis of the REOLYSIN L1 genome segment showed close evolutionary relationship with its human homologs, serotypes Lang and Dearing.


Assuntos
Reoviridae/genética , Genoma Viral/genética , Sequenciamento de Nucleotídeos em Larga Escala , Filogenia , Reoviridae/classificação
8.
Invest New Drugs ; 31(3): 696-706, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22886613

RESUMO

PURPOSE: This open-labeled, phase I clinical trial was designed to determine the safety and tolerability of percutaneous intralesional administration of wild-type oncolytic revovirus type 3 Dearing (Reolysin®) in cancer patients with accessible and evaluable disease, who had otherwise failed to improve on standard cancer interventions. EXPERIMENTAL DESIGN: An escalating dose of Reolysin® starting from up to 10(10) plague forming units (PFU) was administered to each cohort of three patients per dose level. Viral shedding, reovirus neutralizing antibody response, toxicity and clinical response were assessed. RESULTS: Nineteen patients with various advanced solid tumors were treated. The most common toxicities related to treatment were grade 2 (or less) local erythema and transient flu like symptoms. Viral shedding was not seen in cerebral spinal fluid (CSF), urine and stool samples in all patients. Rising viral antibody titres were seen in all patients. In addition, we observed some evidence of local target tumor response activity in 7/19 patients (37 %) at the end of six or more weeks follow-up, with one patient exhibiting a complete response (CR), two a partial response (PR), and four stable disease (SD) to the local injected lesion. CONCLUSIONS: Reolysin® is well tolerated given intralesionaly, with DLT/MTD not reached at a dose of 10(10) PFU. The favorable toxicity profile, lack of viral shedding and possible therapeutic activity has made this unattenuated oncolytic reovirus an attractive cancer therapeutic agent for ongoing clinical studies, including in the setting of locally advanced accessible disease for palliation of symptoms.


Assuntos
Antineoplásicos/administração & dosagem , Orthoreovirus Mamífero 3 , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Administração Cutânea , Adulto , Idoso , Anticorpos Antivirais/sangue , Antineoplásicos/efeitos adversos , Feminino , Cefaleia/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Náusea/etiologia , Neoplasias/sangue , Neoplasias/virologia , Terapia Viral Oncolítica/efeitos adversos , Eliminação de Partículas Virais , Vômito/etiologia
9.
Invest New Drugs ; 31(6): 1476-86, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24121993

RESUMO

Numerous pre-clinical and clinical studies on reovirus have generated valuable information which supports the use of this orphan virus as an investigational drug for cancer treatment. Reolysin® (pelareorep) is a clinical formulation of the human Reovirus Type 3 Dearing strain. The clinical safety and efficacy of Reolysin® in humans is being tested on an assortment of cancer indications as a mono and/or combination therapy. Reovirus has many inherent characteristics that make it a potential candidate for virotherapy, including: the rapid and natural spread through the haematogenous route, the ability to overcome immunological barriers thereby reaching tumor sites, and being replication-competent. The purpose of this study was to elucidate the bio-distribution pattern of Reolysin® in healthy Sprague-Dawley rats. Following a single 15-min intravenous infusion via the tail vein in Sprague-Dawley rats, the levels of virus genome were determined in 16 organs/tissues by RT-qPCR (Reverse Transcriptase- Quantitative Polymerase Chain Reaction) over a 336 h (Day 15) incubation regime. Consistent with previous studies, maximal reovirus RNA levels were observed in the spleen; indicating its involvement in viral uptake and clearance, followed by heart, ovaries, tail (infusion site), liver and lungs. All the organs/tissues demonstrated unquantifiable levels of reovirus genome at the end of incubation, suggesting substantial to complete viral clearance. Several studies in the last decade have described the use of reovirus for treating ovarian cancers. An increase of reovirus genome in ovaries at 24 h post infection was noted. The results will aid in the design of additional exploratory clinical trials for Reolysin®.


Assuntos
Vírus Oncolíticos , RNA Viral/análise , Reoviridae , Animais , Feminino , Infusões Intravenosas , Masculino , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
10.
Biomedicines ; 11(8)2023 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-37626720

RESUMO

Monoclonal antibodies (mAbs) are important treatment modalities for preventing and treating infectious diseases, especially for those lacking prophylactic vaccines or effective therapies. Recent advances in mAb gene cloning from naturally infected or immunized individuals has led to the development of highly potent human mAbs against a wide range of human and animal pathogens. While effective, the serum half-lives of mAbs are quite variable, with single administrations usually resulting in short-term protection, requiring repeated doses to maintain therapeutic concentrations for extended periods of time. Moreover, due to their limited time in circulation, mAb therapies are rarely given prophylactically; instead, they are generally administered therapeutically after the onset of symptoms, thus preventing mortality, but not morbidity. Adeno-associated virus (AAV) vectors have an established record of high-efficiency in vivo gene transfer in a variety of animal models and humans. When delivered to post-mitotic tissues such as skeletal muscle, brain, and heart, or to organs in which cells turn over slowly, such as the liver and lungs, AAV vector genomes assume the form of episomal concatemers that direct transgene expression, often for the lifetime of the cell. Based on these attributes, many research groups have explored AAV-vectored delivery of highly potent mAb genes as a strategy to enable long-term expression of therapeutic mAbs directly in vivo following intramuscular or intranasal administration. However, clinical trials in humans and studies in nonhuman primates (NHPs) indicate that while AAVs are a powerful and promising platform for vectored immunoprophylaxis (VIP), further optimization is needed to decrease anti-drug antibody (ADA) and anti-capsid antibody responses, ultimately leading to increased serum transgene expression levels and improved therapeutic efficacy. The following review will summarize the current landscape of AAV VIP in NHP models, with an emphasis on vector and transgene design as well as general delivery system optimization. In addition, major obstacles to AAV VIP, along with implications for clinical translation, will be discussed.

11.
BMC Cancer ; 12: 368, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22920673

RESUMO

BACKGROUND: Reovirus exploits aberrant signalling downstream of Ras to mediate tumor-specific oncolysis. Since ~90% squamous cell carcinomas of the head and neck (SCCHN) over-express EGFR and SCCHN cell lines are sensitive to oncolytic reovirus, we conducted a detailed analysis of the effects of reovirus in 15 head and neck cancer cell lines. Both pre- and post-entry events were studied in an attempt to define biomarkers predictive of sensitivity/resistance to reovirus. In particular, we analysed the role of EGFR/Ras signalling in determining virus-mediated cytotoxicity in SCCHN. METHODS: To test whether EGFR pathway activity was predictive of increased sensitivity to reovirus, correlative analyses between reoviral IC50 by MTT assay and EGFR levels by western blot and FACS were conducted. Inhibition or stimulation of EGFR signalling were analysed for their effect on reoviral oncolysis by MTT assay, and viral growth by TCID50 assay. We next analysed the effects of inhibiting signalling downstream of Ras, by specific inhibitors of p38MAPK, PI3-K or MEK, on reoviral killing examined by MTT assay. The role of PKR in reoviral killing was also determined by blockade of PKR using 2-aminopurine and assaying for cell survival by MTT assay. The apoptotic response of SCCHN to reovirus was examined by western blot analysis of caspase 3 cleavage. RESULTS: Correlative analyses between reoviral sensitivity and EGFR levels revealed no association. Intermediate sub-viral and core particles showed the same infectivity/cytotoxicity as intact reovirus. Therefore, sensitivity was not determined by cell entry. In 4 cell lines, oncolysis and viral growth were both unaffected by inhibition or stimulation of EGFR signalling. Inhibition of signalling downstream of Ras did not abrogate reoviral oncolysis and, in addition, modulation of PKR using 2-aminopurine did not alter reovirus sensitivity in resistant cell lines. Caspase 3 cleavage was not detected in infected cells and oncolysis was observed in pan-caspase inhibited cells. CONCLUSIONS: In summary, reovirus is potently oncolytic in a broad panel of SCCHN cell lines. Attempts to define sensitivity/resistance by analysis of the EGFR/Ras/MAPK pathway have failed to provide a clear predictive biomarker of response. Further analysis of material from in vitro and clinical studies is ongoing in an attempt to shed further light on this issue.


Assuntos
Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/virologia , Vírus Oncolíticos/metabolismo , Infecções por Reoviridae/metabolismo , Transdução de Sinais/fisiologia , Western Blotting , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Citometria de Fluxo , Humanos , Reoviridae
12.
Biomedicines ; 10(12)2022 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-36551918

RESUMO

Pseudomonas aeruginosa is a bacterial pathogen of global concern and is responsible for 10-15% of nosocomial infections worldwide. This opportunistic bacterial pathogen is known to cause serious complications in immunocompromised patients and is notably the leading cause of morbidity and mortality in patients suffering from cystic fibrosis. Currently, the only line of defense against P. aeruginosa infections is antibiotic treatment. Due to the acquired and adaptive resistance mechanisms of this pathogen, the prevalence of multidrug resistant P. aeruginosa strains has increased, presenting a major problem in healthcare settings. To date, there are no approved licensed vaccines to protect against P. aeruginosa infections, prompting the urgent need alternative treatment options. An alternative to traditional vaccines is vectored immunoprophylaxis (VIP), which utilizes a safe and effective adeno-associated virus (AAV) gene therapy vector to produce sustained levels of therapeutic monoclonal antibodies (mAbs) in vivo from a single intramuscular injection. In this review, we will provide an overview of P. aeruginosa biology and key mechanisms of pathogenesis, discuss current and emerging treatment strategies for P. aeruginosa infections and highlight AAV-VIP as a promising novel therapeutic platform.

13.
Mol Ther Methods Clin Dev ; 26: 505-518, 2022 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-36092367

RESUMO

Filoviruses cause severe hemorrhagic fever with case fatality rates as high as 90%. Filovirus-specific monoclonal antibodies (mAbs) confer protection in nonhuman primates as late as 5 days after challenge, and FDA-approved mAbs REGN-EB3 and mAb114 have demonstrated efficacy against Ebola virus (EBOV) infection in humans. Vectorized antibody expression mediated by adeno-associated virus (AAV) can generate protective and sustained concentrations of therapeutic mAbs in animal models for a variety of infectious diseases, including EBOV. Here we demonstrate that AAV6.2FF-mediated expression of murine IgG2a EBOV mAbs, 2G4 and 5D2, protects from mouse-adapted (MA)-EBOV infection with none of the surviving mice developing anti-VP40 antibodies above background. Protective serum concentrations of AAV6.2FF-2G4/AAV6.2FF-5D2 did not alter endogenous antibody responses to heterologous virus infection. AAV-mediated expression of EBOV mAbs 100 and 114, and pan-ebolavirus mAbs, FVM04, ADI-15878, and CA45, as human IgG1 antibodies conferred protection against MA-EBOV at low serum concentrations, with minimum protective serum levels as low as 2 µg/mL. Vectorized expression of murine IgG2a or human IgG1 mAbs led to sustained expression in the serum of mice for >400 days or for the lifetime of the animal, respectively. AAV6.2FF-mediated mAb expression offers an alternative to recombinant antibody administration in scenarios where long-term protection is preferable to passive immunization.

14.
Biomedicines ; 10(2)2022 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-35203573

RESUMO

Epithelial ovarian cancer is the deadliest gynecological malignancy. The lack of effective treatments highlights the need for novel therapeutic interventions. The aim of this study was to investigate whether sustained adeno-associated virus (AAV) vector-mediated expression of vascular normalizing agents 3TSR and Fc3TSR and the antiangiogenic monoclonal antibody, Bevacizumab, with or without oncolytic virus treatment would improve survival in an orthotopic syngeneic mouse model of epithelial ovarian carcinoma. AAV vectors were administered 40 days post-tumor implantation and combined with oncolytic avian orthoavulavirus-1 (AOaV-1) 20 days later, at the peak of AAV-transgene expression, to ascertain whether survival could be extended. Flow cytometry conducted on blood samples, taken at an acute time point post-AOaV-1 administration (36 h), revealed a significant increase in activated NK cells in the blood of all mice that received AOaV-1. T cell analysis revealed a significant increase in CD8+ tumor specific T cells in the blood of AAV-Bevacizumab+AOaV-1 treated mice compared to control mice 10 days post AOaV-1 administration. Immunohistochemical staining of primary tumors harvested from a subset of mice euthanized 90 days post tumor implantation, when mice typically have large primary tumors, secondary peritoneal lesions, and extensive ascites fluid production, revealed that AAV-3TSR, AAV-Fc3TSR+AOaV-1, or AAV-Bevacizumab+AOaV-1 treated mice had significantly more tumor-infiltrating CD8+ T cells than PBS controls. Despite AAV-mediated transgene expression waning faster in tumor-bearing mice than in non-tumor bearing mice, all three of the AAV therapies significantly extended survival compared to control mice; with AAV-Bevacizumab performing the best in this model. However, combining AAV therapies with a single dose of AOaV-1 did not lead to significant extensions in survival compared to AAV therapies on their own, suggesting that additional doses of AOaV-1 may be required to improve efficacy in this model. These results suggest that vectorizing anti-angiogenic and vascular normalizing agents is a viable therapeutic option that warrants further investigation, including optimizing combination therapies.

15.
Biomedicines ; 9(9)2021 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-34572372

RESUMO

Adeno-associated virus (AAV) vector mediated expression of therapeutic monoclonal antibodies is an alternative strategy to traditional vaccination to generate immunity in immunosuppressed or immunosenescent individuals. In this study, we vectorized a human monoclonal antibody (31C2) directed against the spike protein of SARS-CoV-2 and determined the safety profile of this AAV vector in mice and sheep as a large animal model. In both studies, plasma biochemical parameters and hematology were comparable to untreated controls. Except for mild myositis at the site of injection, none of the major organs revealed any signs of toxicity. AAV-mediated human IgG expression increased steadily throughout the 28-day study in sheep, resulting in peak concentrations of 21.4-46.7 µg/ mL, demonstrating practical scale up from rodent to large animal models. This alternative approach to immunity is worth further exploration after this demonstration of safety, tolerability, and scalability in a large animal model.

16.
Ecol Appl ; 20(2): 465-82, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20405800

RESUMO

Ecosystem models have been developed for assessment and management in a wide variety of environments. As model complexity increases, it becomes more difficult to trace how imperfect knowledge of internal model parameters, data inputs, or relationships among parameters might impact model results, affecting predictions and subsequent management decisions. Sensitivity analysis is an essential component of model evaluation, particularly when models are used to make management decisions. Results should be expressed as probabilities and should realistically account for uncertainty. When models are particularly complex, this can be difficult to do and to present in ways that do not obfuscate essential results. We conducted a sensitivity analysis of the Ecosystem Diagnosis and Treatment (EDT) model, which predicts salmon productivity and capacity as a function of ecosystem conditions. We used a novel "structured sensitivity analysis" approach that is particularly useful for very complex models or those with an abundance of interconnected parameters. We identified small, medium, and large plausible ranges for both input data and model parameters. Using a Monte Carlo approach, we explored the variation in output, prediction intervals, and sensitivity indices, given these plausible input distributions. The analyses indicated that, as a consequence of internal parameter uncertainty, EDT productivity and capacity predictions lack the precision needed for many management applications. However, EDT prioritization of reaches for preservation or restoration was more robust to given input uncertainties, indicating that EDT may be more useful as a relative measure of fish performance than as an absolute measure. Like all large models, if EDT output is to be used as input to other models or management tools it is important to explicitly incorporate the uncertainty and sensitivity analyses into such secondary analyses. Sensitivity analyses should become standard operating procedure for evaluation of ecosystem models.


Assuntos
Ecossistema , Monitoramento Ambiental/métodos , Modelos Teóricos , Salmão , Incerteza , Animais
17.
Clin Cancer Res ; 14(3): 912-23, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18245555

RESUMO

PURPOSE: To test combination treatment schedules of reovirus and radiation in human and murine tumor cells in vitro and in vivo. EXPERIMENTAL DESIGN: In vitro cytotoxicity and cell cycle effects of reovirus given alone and combined with radiotherapy were assessed by colorimetric, tissue culture infectious dose 50, and fluorescence-activated cell sorting-based assays. Interactions between the agents were evaluated using combination index analysis. The effect of different schedules of reovirus and radiotherapy on viral replication and cytotoxicity was tested in vitro and the combination was assessed in three tumor models in vivo. RESULTS: Characterization of reovirus cytotoxicity in a panel of cell lines yielded a range of sensitivities. Combined reovirus and radiotherapy yielded statistically significantly increased cytotoxicity, particularly in cell lines with moderate susceptibility to reovirus alone. The enhanced cytotoxicity of the combination occurred independently of treatment sequence or schedule. Radiation did not affect viral replication and only reduced reoviral cytotoxicity after clinically irrelevant single doses (>50 Gy). Combination index analysis revealed synergy between radiation (3-10 Gy) and reovirus at multiplicities of infection between 0.001 and 1. Combination treatment significantly increased apoptosis in tumor cells relative to either single-agent treatment. In vivo studies using xenograft and syngeneic tumors showed enhanced activity of the combination relative to reovirus or radiation alone (P < 0.001). CONCLUSIONS: Combining reovirus and radiotherapy synergistically enhances cytotoxicity in a variety of tumor cells in vitro and in vivo. These results offer strong support for translational clinical trials of reovirus plus radiotherapy that have been initiated in the clinic.


Assuntos
Sobrevivência Celular/efeitos da radiação , Neoplasias/virologia , Reoviridae/patogenicidade , Adulto , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Neoplasias Colorretais/radioterapia , Neoplasias Colorretais/virologia , Terapia Combinada , Vírus da Dengue , Neoplasias de Cabeça e Pescoço/radioterapia , Neoplasias de Cabeça e Pescoço/virologia , Humanos , Raios X
18.
Mol Ther ; 16(3): 627-32, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18253152

RESUMO

Reovirus is an oncolytic virus with activity in in vivo models of malignant gliomas (MGs). The primary aims were to determine the dose-limiting toxicity (DLT) and maximum tolerated dose (MTD) of intratumoral administration of reovirus in patients with recurrent MGs. Response, survival, and time to progression (TTP) were secondary aims. Patients were adults, had Karnofsky Performance score > or = 60, received prior radiotherapy with or without chemotherapy, and had up to the third recurrence of MG. Reovirus was administered intratumorally stereotactically at 1 x 10(7), 1 x 10(8), or 1 x 10(9) tissue culture infectious dose 50 (TCID50) in a volume of 0.9 ml. Twelve patients were treated at three dose levels (3, 6, and 3 patients, respectively). Seven were men, median Karnofsky Performance score was 80, and median age was 53.5 years. There were no grade III or IV adverse events (AEs) definitely or probably related to treatment. Ten patients had tumor progression, one had stabilization, and one was not evaluable for response. Median survival was 21 weeks (range, 6-234), and one is alive 54 months after treatment. Median TTP was 4.3 weeks (range, 2.6-39). An MTD was not reached. The intratumoral administration of the genetically unmodified reovirus was well tolerated using these doses and schedule, in patients with recurrent MG.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Terapia Viral Oncolítica/métodos , Reoviridae/fisiologia , Adulto , Anticorpos Antivirais/sangue , Neoplasias Encefálicas/patologia , Feminino , Glioma/patologia , Cefaleia/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Debilidade Muscular/etiologia , Recidiva Local de Neoplasia , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos/imunologia , Vírus Oncolíticos/fisiologia , Reoviridae/imunologia , Análise de Sobrevida , Resultado do Tratamento
19.
Acad Radiol ; 15(5): 571-5, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18423313

RESUMO

RATIONALE AND OBJECTIVES: We sought to assess the performance of a real-time interactive pulmonary nodule analysis system for evaluation of chest digital radiographic (DR) images in a routine clinical environment. MATERIALS AND METHODS: A real-time interactive pulmonary nodule analysis system for chest DR image softcopy reading (IQQA-Chest; EDDA Technology, Princeton Junction, NJ) was used in daily practice with a Picture Archiving and Communication System in a National Cancer Institute-designated cancer teaching hospital. Patients referred for follow-up of known cancer underwent digital chest radiography. Posteroanterior and lateral DR images were first read by resident radiologists along with experienced chest radiologists using a Picture Archiving and Communication System work station. The computer-assisted detection (CAD) program was subsequently applied to the posteroanterior DR images, and changes (if any) in diagnosis were recorded. For reference standard, a follow-up chest radiograph at least 6 months following the initial examination or a follow-up computed tomographic scan of the chest within 3 months was used to establish diagnostic accuracy. RESULTS: Of 324 DR examinations, follow-up imaging according to our parameters was available for 214 patients (67%). Lung nodules were found and subsequently confirmed in 35 patients (10%) without CAD. Using CAD, nodules were found and subsequently confirmed in 51 patients (15%), improving sensitivity from 63.8% (95% confidence interval [CI], 0.49%-0.76%) to 92.7% (95% CI, 0.82%-0.98%) (P < .0001, McNemar). Nodules were subsequently proved to be malignant in five of the 16 additional cases (31%). False-positive readings increased from three to six cases; specificity decreased from 98.1% (95% CI, 0.95%-0.99%) to 96.2% (95% CI, 0.92%-0.98%) (not significant). There were 153 true-negative cases (71.4%). CONCLUSIONS: This study suggests that the interpretation of chest radiographs for lung nodules can be improved using an automated CAD nodule detection system. This improvement in reader performance comes with a minimal number of false-positive interpretations.


Assuntos
Diagnóstico por Computador/instrumentação , Neoplasias Pulmonares/diagnóstico por imagem , Interpretação de Imagem Radiográfica Assistida por Computador/instrumentação , Radiografia Torácica , Nódulo Pulmonar Solitário/diagnóstico por imagem , Distribuição de Qui-Quadrado , Humanos , Neoplasias Pulmonares/secundário , Valor Preditivo dos Testes , Estudos Prospectivos , Sistemas de Informação em Radiologia , Sensibilidade e Especificidade , Software , Nódulo Pulmonar Solitário/secundário , Interface Usuário-Computador
20.
Acad Radiol ; 15(7): 835-43, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18572118

RESUMO

RATIONALE AND OBJECTIVES: Evaluate the role of two-dimensional echocardiography and electrocardiographically (ECG)-gated contrast-enhanced multislice computed tomographic (MSCT) cardiac imaging to assess cardiac anatomy, specifically pulmonary venous anatomy and left atrial thrombus, in a selected group of patients before catheter-based atrial fibrillation ablation. MATERIALS AND METHODS: Left atrial anatomy and associated findings in 34 consecutive patients scheduled for electrophysiologic testing who underwent both echocardiography and ECG-gated 16-slice MSCT cardiac imaging were retrospectively compared. Results from two-dimensional transthoracic echocardiography (TTE), cardiac MSCT, electrophysiologic study (EPS), and transesophageal echocardiography (TEE) (when performed) were taken from the official medical record without prior knowledge of this study when interpretation was rendered for clinical use. Electronic record review included: presence of left atrial thrombus (defined as constant filling defect on at least two echocardiographic views or filling defect on computed tomography) and location, pulmonary venous anatomy, and other cardiac, mediastinal, or pulmonary abnormalities. RESULTS: Left atrial thrombus was identified by cardiac MSCT alone in five patients (15%). Pulmonary venous variants were identified with cardiac MSCT in two patients (6%). Both MSCT and echocardiography were normal in 17 subjects (79%). Echocardiography was better at identifying associated valvular abnormalities that were seen in 10 patients (29%). Cardiac MSCT angiography alone identified other cardiac and noncardiac abnormalities, including suspicious pulmonary malignancy, mediastinal adenopathy, and coronary stenosis in 15 patients (44%). CONCLUSIONS: Echocardiography and cardiac MSCT angiography often provide complimentary findings during the preprocedural evaluation for patients with atrial fibrillation requiring ablation. Cardiac MSCT may provide significant additional information about the left atrium, mediastinum, coronary circulation, and visualized lung fields. Based on this study, we would advise that patients considered for radiofrequency ablation for uncontrolled right atrial fibrillation have both echocardiography and ECG-gated contrast-enhanced cardiac MSCT performed as part of the preprocedure evaluation.


Assuntos
Fibrilação Atrial/diagnóstico por imagem , Ablação por Cateter , Trombose Coronária/diagnóstico por imagem , Ecocardiografia , Átrios do Coração/diagnóstico por imagem , Veias Pulmonares/diagnóstico por imagem , Tomografia Computadorizada por Raios X , Idoso , Fibrilação Atrial/cirurgia , Meios de Contraste , Trombose Coronária/cirurgia , Eletrocardiografia , Feminino , Átrios do Coração/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Cuidados Pré-Operatórios , Veias Pulmonares/anormalidades , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA