26 results on '"Aschenbrenner L"'
Search Results
2. Biomarkers for Diagnosis of Sepsis in Patients with Liver Cirrhosis
- Author
-
Pavel, V, additional, Mester, P, additional, Gülow, K, additional, Kunst, C, additional, Aschenbrenner, L, additional, Müller-Schilling, M, additional, and Schmid, S, additional
- Published
- 2021
- Full Text
- View/download PDF
3. Finite pointset method (FPM): optimized meshfree solver for airbag deployment simulations
- Author
-
Jörg Kuhnert, Aschenbrenner, L., and Trameçon, A.
- Subjects
Finite element method ,Computational methods in mechanics ,Particle methods (Numerical analysis) ,Meshfree Methods, General Finite Difference Scheme, Airbag Deployment ,Elements finits, Mètode dels ,Matemàtiques i estadística::Anàlisi numèrica::Mètodes en elements finits [Àrees temàtiques de la UPC] - Abstract
This document provides an outline of the complete numerical scheme of the Finite Pointset Method (FPM), a meshfree general Finite Difference idea to simulate gasdynamics. The method uses a specialized upwind formulation as well as specialized discrete numerical differential operators in order to gain accuracy as well as stability. The scheme is completed by a k-epsilon turbulence model in order to account for viscous effects. Industrially, FPM performs in airbag deployment modelling as a part of full crash simulations in vehicle design and development.
- Published
- 2011
4. Comment on: Concerns of using sialidase fusion protein as an experimental drug to combat seasonal and pandemic influenza
- Author
-
Nicholls, J. M., primary, Aschenbrenner, L. M., additional, Paulson, J. C., additional, Campbell, E. R., additional, Malakhov, M. P., additional, Wurtman, D. F., additional, Yu, M., additional, and Fang, F., additional
- Published
- 2008
- Full Text
- View/download PDF
5. Ornithologische Mitteilungen : Monatsschrift für Vogelbeobachtung, Feldornithologie und Avifaunistik / von Thiede, Walther / Sturmmöwen (Larus canus) als Nahrungsschmarotzer bei Lachmöwen
- Author
-
Peters, Hanns, Aschenbrenner, L., Peters, Hanns, and Aschenbrenner, L.
- Abstract
Hanns Peters, Wien ; L. Aschenbrenner, Wien
- Published
- 1958
6. Discovery of SARS-CoV-2 papain-like protease (PL pro ) inhibitors with efficacy in a murine infection model.
- Author
-
Garnsey MR, Robinson MC, Nguyen LT, Cardin R, Tillotson J, Mashalidis E, Yu A, Aschenbrenner L, Balesano A, Behzadi A, Boras B, Chang JS, Eng H, Ephron A, Foley T, Ford KK, Frick JM, Gibson S, Hao L, Hurst B, Kalgutkar AS, Korczynska M, Lengyel-Zhand Z, Gao L, Meredith HR, Patel NC, Polivkova J, Rai D, Rose CR, Rothan H, Sakata SK, Vargo TR, Qi W, Wu H, Liu Y, Yurgelonis I, Zhang J, Zhu Y, Zhang L, and Lee AA
- Subjects
- Animals, Mice, Humans, COVID-19 virology, Protease Inhibitors pharmacology, Protease Inhibitors chemistry, Protease Inhibitors therapeutic use, Coronavirus 3C Proteases antagonists & inhibitors, Coronavirus 3C Proteases metabolism, Machine Learning, Female, Virus Replication drug effects, SARS-CoV-2 drug effects, COVID-19 Drug Treatment, Antiviral Agents pharmacology, Antiviral Agents chemistry, Antiviral Agents pharmacokinetics, Antiviral Agents therapeutic use, Disease Models, Animal, Coronavirus Papain-Like Proteases antagonists & inhibitors, Coronavirus Papain-Like Proteases metabolism
- Abstract
Vaccines and first-generation antiviral therapeutics have provided important protection against COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, there remains a need for additional therapeutic options that provide enhanced efficacy and protection against potential viral resistance. The SARS-CoV-2 papain-like protease (PL
pro ) is one of the two essential cysteine proteases involved in viral replication. While inhibitors of the SARS-CoV-2 main protease have demonstrated clinical efficacy, known PLpro inhibitors have, to date, lacked the inhibitory potency and requisite pharmacokinetics to demonstrate that targeting PLpro translates to in vivo efficacy in a preclinical setting. Here, we report the machine learning-driven discovery of potent, selective, and orally available SARS-CoV-2 PLpro inhibitors, with lead compound PF-07957472 ( 4 ) providing robust efficacy in a mouse-adapted model of COVID-19 infection.- Published
- 2024
- Full Text
- View/download PDF
7. Generation of a VeroE6 Pgp gene knock out cell line and its use in SARS-CoV-2 antiviral study.
- Author
-
Zhu Y, Binder J, Yurgelonis I, Rai DK, Lazarro S, Costales C, Kobylarz K, McMonagle P, Steppan CM, Aschenbrenner L, Anderson AS, and Cardin RD
- Subjects
- Humans, Chlorocebus aethiops, Animals, Antiviral Agents pharmacology, Gene Knockout Techniques, Vero Cells, ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Cell Line, SARS-CoV-2 genetics, COVID-19 Drug Treatment
- Abstract
Vero cells are widely used for antiviral tests and virology research for SARS-CoV-2 as well as viruses from various other families. However, Vero cells generally express high levels of multi-drug resistance 1 (MDR1) or Pgp protein, the efflux transporter of foreign substances including many antiviral compounds, affecting the antiviral activity as well as interpretation of data. To address this, a Pgp gene knockout VeroE6 cell line (VeroE6-Pgp-KO) was generated using CRISPR-CAS9 technology. These cells no longer expressed the Pgp protein as indicated by flow cytometry analysis following staining with a Pgp-specific monoclonal antibody. They also showed significantly reduced efflux transporter activity in the calcein acetoxymethyl ester (calcein AM) assay. The VeroE6-Pgp-KO cells and the parental VeroE6 cells were each infected with SARS-CoV-2 to test antiviral activities of remdesivir and nirmatrelvir, two known Pgp substrates, in the presence or absence of a Pgp inhibitor. The compounds showed antiviral activities in VeroE6-Pgp-KO cells similar to that observed in the presence of the Pgp inhibitor. Thus, the newly established VeroE6-Pgp-KO cell line adds a new in vitro virus infection system for SARS-CoV-2 and possibly other viruses to test antiviral therapies without a need to control the Pgp activity. Removal of the Pgp inhibitor for antiviral assays will lead to less data variation and prevent failed assays., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: All authors are employees of Pfizer, Inc., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
8. An oral SARS-CoV-2 M pro inhibitor clinical candidate for the treatment of COVID-19.
- Author
-
Owen DR, Allerton CMN, Anderson AS, Aschenbrenner L, Avery M, Berritt S, Boras B, Cardin RD, Carlo A, Coffman KJ, Dantonio A, Di L, Eng H, Ferre R, Gajiwala KS, Gibson SA, Greasley SE, Hurst BL, Kadar EP, Kalgutkar AS, Lee JC, Lee J, Liu W, Mason SW, Noell S, Novak JJ, Obach RS, Ogilvie K, Patel NC, Pettersson M, Rai DK, Reese MR, Sammons MF, Sathish JG, Singh RSP, Steppan CM, Stewart AE, Tuttle JB, Updyke L, Verhoest PR, Wei L, Yang Q, and Zhu Y
- Subjects
- Administration, Oral, Animals, COVID-19 virology, Clinical Trials, Phase I as Topic, Coronavirus drug effects, Disease Models, Animal, Drug Therapy, Combination, Humans, Lactams administration & dosage, Lactams pharmacokinetics, Leucine administration & dosage, Leucine pharmacokinetics, Mice, Mice, Inbred BALB C, Microbial Sensitivity Tests, Nitriles administration & dosage, Nitriles pharmacokinetics, Proline administration & dosage, Proline pharmacokinetics, Randomized Controlled Trials as Topic, Ritonavir administration & dosage, Ritonavir therapeutic use, SARS-CoV-2 physiology, Viral Protease Inhibitors administration & dosage, Viral Protease Inhibitors pharmacokinetics, Virus Replication drug effects, Lactams pharmacology, Lactams therapeutic use, Leucine pharmacology, Leucine therapeutic use, Nitriles pharmacology, Nitriles therapeutic use, Proline pharmacology, Proline therapeutic use, SARS-CoV-2 drug effects, Viral Protease Inhibitors pharmacology, Viral Protease Inhibitors therapeutic use, COVID-19 Drug Treatment
- Abstract
The worldwide outbreak of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic. Alongside vaccines, antiviral therapeutics are an important part of the healthcare response to countering the ongoing threat presented by COVID-19. Here, we report the discovery and characterization of PF-07321332, an orally bioavailable SARS-CoV-2 main protease inhibitor with in vitro pan-human coronavirus antiviral activity and excellent off-target selectivity and in vivo safety profiles. PF-07321332 has demonstrated oral activity in a mouse-adapted SARS-CoV-2 model and has achieved oral plasma concentrations exceeding the in vitro antiviral cell potency in a phase 1 clinical trial in healthy human participants.
- Published
- 2021
- Full Text
- View/download PDF
9. Preclinical characterization of an intravenous coronavirus 3CL protease inhibitor for the potential treatment of COVID19.
- Author
-
Boras B, Jones RM, Anson BJ, Arenson D, Aschenbrenner L, Bakowski MA, Beutler N, Binder J, Chen E, Eng H, Hammond H, Hammond J, Haupt RE, Hoffman R, Kadar EP, Kania R, Kimoto E, Kirkpatrick MG, Lanyon L, Lendy EK, Lillis JR, Logue J, Luthra SA, Ma C, Mason SW, McGrath ME, Noell S, Obach RS, O' Brien MN, O'Connor R, Ogilvie K, Owen D, Pettersson M, Reese MR, Rogers TF, Rosales R, Rossulek MI, Sathish JG, Shirai N, Steppan C, Ticehurst M, Updyke LW, Weston S, Zhu Y, White KM, García-Sastre A, Wang J, Chatterjee AK, Mesecar AD, Frieman MB, Anderson AS, and Allerton C
- Subjects
- Adenosine Monophosphate administration & dosage, Adenosine Monophosphate adverse effects, Adenosine Monophosphate analogs & derivatives, Adenosine Monophosphate pharmacokinetics, Alanine administration & dosage, Alanine adverse effects, Alanine analogs & derivatives, Alanine pharmacokinetics, Animals, COVID-19 virology, Chlorocebus aethiops, Coronavirus 229E, Human drug effects, Coronavirus 229E, Human enzymology, Coronavirus Protease Inhibitors adverse effects, Coronavirus Protease Inhibitors pharmacokinetics, Disease Models, Animal, Drug Design, Drug Synergism, Drug Therapy, Combination, HeLa Cells, Humans, Indoles adverse effects, Indoles pharmacokinetics, Infusions, Intravenous, Leucine adverse effects, Leucine pharmacokinetics, Mice, Pyrrolidinones adverse effects, Pyrrolidinones pharmacokinetics, Severe acute respiratory syndrome-related coronavirus drug effects, Severe acute respiratory syndrome-related coronavirus enzymology, SARS-CoV-2 drug effects, SARS-CoV-2 enzymology, Vero Cells, Coronavirus 3C Proteases antagonists & inhibitors, Coronavirus Protease Inhibitors administration & dosage, Indoles administration & dosage, Leucine administration & dosage, Pyrrolidinones administration & dosage, COVID-19 Drug Treatment
- Abstract
COVID-19 caused by the SARS-CoV-2 virus has become a global pandemic. 3CL protease is a virally encoded protein that is essential across a broad spectrum of coronaviruses with no close human analogs. PF-00835231, a 3CL protease inhibitor, has exhibited potent in vitro antiviral activity against SARS-CoV-2 as a single agent. Here we report, the design and characterization of a phosphate prodrug PF-07304814 to enable the delivery and projected sustained systemic exposure in human of PF-00835231 to inhibit coronavirus family 3CL protease activity with selectivity over human host protease targets. Furthermore, we show that PF-00835231 has additive/synergistic activity in combination with remdesivir. We present the ADME, safety, in vitro, and in vivo antiviral activity data that supports the clinical evaluation of PF-07304814 as a potential COVID-19 treatment., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
10. Acoustic Ejection Mass Spectrometry for High-Throughput Analysis.
- Author
-
Zhang H, Liu C, Hua W, Ghislain LP, Liu J, Aschenbrenner L, Noell S, Dirico KJ, Lanyon LF, Steppan CM, West M, Arnold DW, Covey TR, Datwani SS, and Troutman MD
- Subjects
- Acoustics, High-Throughput Screening Assays, Spectrometry, Mass, Electrospray Ionization
- Abstract
We describe a mass spectrometry (MS) analytical platform resulting from the novel integration of acoustic droplet ejection (ADE) technology, an open-port interface (OPI), and electrospray ionization (ESI)-MS that creates a transformative system enabling high-speed sampling and label-free analysis. The ADE technology delivers nanoliter droplets in a touchless manner with high speed, precision, and accuracy. Subsequent sample dilution within the OPI, in concert with the capabilities of modern ESI-MS, eliminates the laborious sample preparation and method development required in current approaches. This platform is applied to a variety of experiments, including high-throughput (HT) pharmacology screening, label-free in situ enzyme kinetics, in vitro absorption, distribution, metabolism, elimination, pharmacokinetic and biomarker analysis, and HT parallel medicinal chemistry.
- Published
- 2021
- Full Text
- View/download PDF
11. Discovery of a Novel Inhibitor of Coronavirus 3CL Protease for the Potential Treatment of COVID-19.
- Author
-
Boras B, Jones RM, Anson BJ, Arenson D, Aschenbrenner L, Bakowski MA, Beutler N, Binder J, Chen E, Eng H, Hammond H, Hammond J, Haupt RE, Hoffman R, Kadar EP, Kania R, Kimoto E, Kirkpatrick MG, Lanyon L, Lendy EK, Lillis JR, Logue J, Luthra SA, Ma C, Mason SW, McGrath ME, Noell S, Obach RS, O'Brien MN, O'Connor R, Ogilvie K, Owen D, Pettersson M, Reese MR, Rogers TF, Rossulek MI, Sathish JG, Shirai N, Steppan C, Ticehurst M, Updyke LW, Weston S, Zhu Y, Wang J, Chatterjee AK, Mesecar AD, Frieman MB, Anderson AS, and Allerton C
- Abstract
COVID-19 caused by the SARS-CoV-2 virus has become a global pandemic. 3CL protease is a virally encoded protein that is essential across a broad spectrum of coronaviruses with no close human analogs. The designed phosphate prodrug PF-07304814 is metabolized to PF-00835321 which is a potent inhibitor in vitro of the coronavirus family 3CL pro, with selectivity over human host protease targets. Furthermore, PF-00835231 exhibits potent in vitro antiviral activity against SARS-CoV-2 as a single agent and it is additive/synergistic in combination with remdesivir. We present the ADME, safety, in vitro , and in vivo antiviral activity data that supports the clinical evaluation of this compound as a potential COVID-19 treatment.
- Published
- 2021
- Full Text
- View/download PDF
12. Nonclinical safety assessment of epigenetic modulatory drugs: Current status and industry perspective.
- Author
-
Reynolds VL, Butler P, Abernathy MM, Aschenbrenner L, Best DD, Blank J, Crosby M, Custer L, Escobar PA, Kolaja K, Moggs J, Shuey D, Snyder C, Van Vleet T, Zhou J, and Hart TK
- Subjects
- Animals, Drug Evaluation, Preclinical standards, Drug Evaluation, Preclinical trends, Drug Industry trends, Drug-Related Side Effects and Adverse Reactions epidemiology, Drug-Related Side Effects and Adverse Reactions prevention & control, Epigenesis, Genetic genetics, Humans, Pharmaceutical Preparations administration & dosage, Risk Assessment standards, Risk Assessment trends, Drug Industry standards, Drug and Narcotic Control trends, Epigenesis, Genetic drug effects, Pharmaceutical Preparations standards, Surveys and Questionnaires
- Abstract
Pharmaceutic products designed to perturb the function of epigenetic modulators have been approved by regulatory authorities for treatment of advanced cancer. While the predominant effort in epigenetic drug development continues to be in oncology, non-oncology indications are also garnering interest. A survey of pharmaceutical companies was conducted to assess the interest and concerns for developing small molecule direct epigenetic effectors (EEs) as medicines. Survey themes addressed (1) general levels of interest and activity with EEs as therapeutic agents, (2) potential safety concerns, and (3) possible future efforts to develop targeted strategies for nonclinical safety assessment of EEs. Thirteen companies contributed data to the survey. Overall, the survey data indicate the consensus opinion that existing ICH guidelines are effective and appropriate for nonclinical safety assessment activities with EEs. Attention in the framework of study design should, on a case by case basis, be considered for delayed or latent toxicities, carcinogenicity, reproductive toxicity, and the theoretical potential for transgenerational effects. While current guidelines have been appropriate for the nonclinical safety assessments of epigenetic targets, broader experience with a wide range of epigenetic targets will provide information to assess the potential need for new or revised risk assessment strategies for EE drugs., (Copyright © 2020. Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
13. Author Correction: Targeting CLDN18.2 by CD3 Bispecific and ADC Modalities for the Treatments of Gastric and Pancreatic Cancer.
- Author
-
Zhu G, Foletti D, Liu X, Ding S, Melton Witt J, Hasa-Moreno A, Rickert M, Holz C, Aschenbrenner L, Yang AH, Kraynov E, Evering W, Obert L, Lee C, Sai T, Mistry T, Lindquist KC, Van Blarcom T, Strop P, Chaparro-Riggers J, and Liu SH
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
14. Preclinical Efficacy and Safety Comparison of CD3 Bispecific and ADC Modalities Targeting BCMA for the Treatment of Multiple Myeloma.
- Author
-
Panowski SH, Kuo TC, Zhang Y, Chen A, Geng T, Aschenbrenner L, Kamperschroer C, Pascua E, Chen W, Delaria K, Farias S, Bateman M, Dushin RG, Chin SM, Van Blarcom TJ, Yeung YA, Lindquist KC, Chunyk AG, Kuang B, Han B, Mirsky M, Pardo I, Buetow B, Martin TG, Wolf JL, Shelton D, Rajpal A, Strop P, Chaparro-Riggers J, and Sasu BJ
- Subjects
- Animals, Antibodies, Bispecific adverse effects, Antibodies, Bispecific pharmacology, Antibody Affinity, B-Cell Maturation Antigen antagonists & inhibitors, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Dose-Response Relationship, Drug, Drug Evaluation, Preclinical, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Immunoconjugates adverse effects, Immunoconjugates pharmacology, Mice, Multiple Myeloma metabolism, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Antibodies, Bispecific administration & dosage, B-Cell Maturation Antigen metabolism, CD3 Complex immunology, Immunoconjugates administration & dosage, Multiple Myeloma drug therapy
- Abstract
The restricted expression pattern of B-cell maturation antigen (BCMA) makes it an ideal tumor-associated antigen (TAA) for the treatment of myeloma. BCMA has been targeted by both CD3 bispecific antibody and antibody-drug conjugate (ADC) modalities, but a true comparison of modalities has yet to be performed. Here we utilized a single BCMA antibody to develop and characterize both a CD3 bispecific and 2 ADC formats (cleavable and noncleavable) and compared activity both in vitro and in vivo with the aim of generating an optimal therapeutic. Antibody affinity, but not epitope was influential in drug activity and hence a high-affinity BCMA antibody was selected. Both the bispecific and ADCs were potent in vitro and in vivo , causing dose-dependent cell killing of myeloma cell lines and tumor regression in orthotopic myeloma xenograft models. Primary patient cells were effectively lysed by both CD3 bispecific and ADCs, with the bispecific demonstrating improved potency, maximal cell killing, and consistency across patients. Safety was evaluated in cynomolgus monkey toxicity studies and both modalities were active based on on-target elimination of B lineage cells. Distinct nonclinical toxicity profiles were seen for the bispecific and ADC modalities. When taken together, results from this comparison of BCMA CD3 bispecific and ADC modalities suggest better efficacy and an improved toxicity profile might be achieved with the bispecific modality. This led to the advancement of a bispecific candidate into phase I clinical trials., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
15. Targeting CLDN18.2 by CD3 Bispecific and ADC Modalities for the Treatments of Gastric and Pancreatic Cancer.
- Author
-
Zhu G, Foletti D, Liu X, Ding S, Melton Witt J, Hasa-Moreno A, Rickert M, Holz C, Aschenbrenner L, Yang AH, Kraynov E, Evering W, Obert L, Lee C, Sai T, Mistry T, Lindquist KC, Van Blarcom T, Strop P, Chaparro-Riggers J, and Liu SH
- Subjects
- Adenocarcinoma therapy, Animals, Carcinoma, Pancreatic Ductal therapy, Cell Line, Tumor, Humans, Immunoconjugates blood, Mice, Pancreatic Neoplasms blood, Rats, Stomach Neoplasms blood, Antibodies, Bispecific immunology, CD3 Complex immunology, Claudins immunology, Immunoconjugates therapeutic use, Pancreatic Neoplasms therapy, Stomach Neoplasms therapy
- Abstract
Human CLDN18.2 is highly expressed in a significant proportion of gastric and pancreatic adenocarcinomas, while normal tissue expression is limited to the epithelium of the stomach. The restricted expression makes it a potential drug target for the treatment of gastric and pancreatic adenocarcinoma, as evidenced by efforts to target CLDN18.2 via naked antibody and CAR-T modalities. Herein we describe CLDN18.2-targeting via a CD3-bispecific and an antibody drug conjugate and the characterization of these potential therapeutic molecules in efficacy and preliminary toxicity studies. Anti-hCLDN18.2 ADC, CD3-bispecific and diabody, targeting a protein sequence conserved in rat, mouse and monkey, exhibited in vitro cytotoxicity in BxPC3/hCLDN18.2 (IC
50 = 1.52, 2.03, and 0.86 nM) and KATO-III/hCLDN18.2 (IC50 = 1.60, 0.71, and 0.07 nM) respectively and inhibited tumor growth of pancreatic and gastric patient-derived xenograft tumors. In a rat exploratory toxicity study, the ADC was tolerated up to 10 mg/kg. In a preliminary assessment of tolerability, the anti-CLDN18.2 diabody (0.34 mg/kg) did not produce obvious signs of toxicity in the stomach of NSG mice 4 weeks after dosing. Taken together, our data indicate that targeting CLDN18.2 with an ADC or bispecific modality could be a valid therapeutic approach for the treatment of gastric and pancreatic cancer.- Published
- 2019
- Full Text
- View/download PDF
16. RN927C, a Site-Specific Trop-2 Antibody-Drug Conjugate (ADC) with Enhanced Stability, Is Highly Efficacious in Preclinical Solid Tumor Models.
- Author
-
Strop P, Tran TT, Dorywalska M, Delaria K, Dushin R, Wong OK, Ho WH, Zhou D, Wu A, Kraynov E, Aschenbrenner L, Han B, O'Donnell CJ, Pons J, Rajpal A, Shelton DL, and Liu SH
- Subjects
- Animals, Antigens, Neoplasm genetics, Antigens, Neoplasm metabolism, Antineoplastic Agents chemistry, Cell Adhesion Molecules genetics, Cell Adhesion Molecules metabolism, Cell Cycle Checkpoints drug effects, Cell Cycle Checkpoints genetics, Cell Line, Tumor, Disease Models, Animal, Drug Stability, Female, Gene Expression, Humans, Immunoconjugates chemistry, Lysosomes, Mice, Mitosis drug effects, Mitosis genetics, Molecular Targeted Therapy, Neoplasms drug therapy, Neoplasms metabolism, Neoplasms pathology, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Cell Adhesion Molecules antagonists & inhibitors, Immunoconjugates pharmacology
- Abstract
Trop-2, also known as TACSTD2, EGP-1, GA733-1, and M1S1, is frequently expressed on a variety of human carcinomas, and its expression is often associated with poor prognosis of the diseases. However, it is also present on the epithelium of several normal tissues. A comprehensively designed Trop-2-targeting antibody-drug conjugate (ADC), balancing both efficacy and toxicity, is therefore necessary to achieve clinical utility. To this end, we developed a cleavable Trop-2 ADC (RN927C) using a site-specific transglutaminase-mediated conjugation method and a proprietary microtubule inhibitor (MTI) linker-payload, PF-06380101. Robust in vitro cytotoxicity of RN927C was observed on a panel of Trop-2-expressing tumor cell lines, with IC
50 generally in the subnanomolar range. As expected for an MTI-containing ADC, RN927C readily induced mitotic arrest of treated cells in vitro and in vivo, followed by subsequent cell death. The in vivo efficacy of RN927C was tested in multiple cell line and patient-derived xenograft tumor models, including pancreatic, lung, ovarian, and triple-negative breast tumor types. Single-dose administration of RN927C at 0.75 to 3 mg/kg was generally sufficient to induce sustained regression of Trop-2-expressing tumors and showed superior efficacy over standard treatment with paclitaxel or gemcitabine. Administration of RN927C in nonhuman primate toxicity studies resulted in target-mediated effects in skin and oral mucosa, consistent with Trop-2 expression in these epithelial tissues with minimal, non-dose limiting off-target toxicities. On the basis of the combined efficacy and safety results, RN927C is postulated to have a favorable therapeutic index for treatment of solid tumors. Mol Cancer Ther; 15(11); 2698-708. ©2016 AACR., (©2016 American Association for Cancer Research.)- Published
- 2016
- Full Text
- View/download PDF
17. Characterization of a respiratory syncytial virus L protein inhibitor.
- Author
-
Tiong-Yip CL, Aschenbrenner L, Johnson KD, McLaughlin RE, Fan J, Challa S, Xiong H, and Yu Q
- Subjects
- Amino Acid Sequence, Cell Line, Cell Survival drug effects, Drug Resistance, Viral, Humans, Molecular Sequence Data, Niacinamide pharmacology, Polymerase Chain Reaction, Replicon genetics, Antiviral Agents pharmacology, Benzazepines pharmacology, Cyclopropanes pharmacology, Niacinamide analogs & derivatives, RNA-Dependent RNA Polymerase antagonists & inhibitors, Respiratory Syncytial Virus Infections virology, Respiratory Syncytial Virus, Human drug effects, Viral Proteins antagonists & inhibitors
- Abstract
The respiratory syncytial virus (RSV) L protein is a viral RNA-dependent RNA polymerase that contains multiple enzyme activities required for RSV replication. The RSV L inhibitors described in literature are limited by their cytotoxicity or the lack of RSV B subtype coverage. Here, we characterize a new RSV L inhibitor with strong antiviral activity against both RSV A and B subtypes and no detectable cytotoxicity. This compound, AZ-27, was equally active against RSV live viruses and subgenomic replicons and demonstrated advantages over other classes of RSV inhibitors in time-of-addition and cell line dependency studies. Resistance studies identified a dominant mutation in the putative capping enzyme domain of L protein, which conferred strong resistance to the AZ-27 series but not other classes of RSV inhibitors, supporting RSV L protein as the direct target for AZ-27. This novel and broad-spectrum RSV L polymerase inhibitor may pave the way toward an efficacious RSV therapeutic and provide a new tool for interrogation of the L protein function., (Copyright © 2014, American Society for Microbiology. All Rights Reserved.)
- Published
- 2014
- Full Text
- View/download PDF
18. Siderophore receptor-mediated uptake of lactivicin analogues in gram-negative bacteria.
- Author
-
Starr J, Brown MF, Aschenbrenner L, Caspers N, Che Y, Gerstenberger BS, Huband M, Knafels JD, Lemmon MM, Li C, McCurdy SP, McElroy E, Rauckhorst MR, Tomaras AP, Young JA, Zaniewski RP, Shanmugasundaram V, and Han S
- Subjects
- Gram-Negative Bacteria drug effects, Gram-Negative Bacteria enzymology, Microbial Sensitivity Tests, Peptides drug effects, Peptides, Cyclic, beta-Lactamases metabolism, Gram-Negative Bacteria metabolism, Peptides metabolism, Receptors, Cell Surface metabolism, Siderophores metabolism
- Abstract
Multidrug-resistant Gram-negative pathogens are an emerging threat to human health, and addressing this challenge will require development of new antibacterial agents. This can be achieved through an improved molecular understanding of drug-target interactions combined with enhanced delivery of these agents to the site of action. Herein we describe the first application of siderophore receptor-mediated drug uptake of lactivicin analogues as a strategy that enables the development of novel antibacterial agents against clinically relevant Gram-negative bacteria. We report the first crystal structures of several sideromimic conjugated compounds bound to penicillin binding proteins PBP3 and PBP1a from Pseudomonas aeruginosa and characterize the reactivity of lactivicin and β-lactam core structures. Results from drug sensitivity studies with β-lactamase enzymes are presented, as well as a structure-based hypothesis to reduce susceptibility to this enzyme class. Finally, mechanistic studies demonstrating that sideromimic modification alters the drug uptake process are discussed.
- Published
- 2014
- Full Text
- View/download PDF
19. Discovery of a potent respiratory syncytial virus RNA polymerase inhibitor.
- Author
-
Xiong H, Foulk M, Aschenbrenner L, Fan J, Tiong-Yip CL, Johnson KD, Moustakas D, Fleming PR, Brown DG, Zhang M, Ferguson D, Wu D, and Yu Q
- Subjects
- Antiviral Agents chemical synthesis, Antiviral Agents chemistry, Antiviral Agents pharmacology, Azepines chemical synthesis, Azepines chemistry, Azepines pharmacology, Cell Line, DNA-Directed RNA Polymerases metabolism, Drug Evaluation, Preclinical, Enzyme Activation drug effects, Enzyme Inhibitors chemical synthesis, Humans, Structure-Activity Relationship, Viral Proteins metabolism, Virus Replication drug effects, DNA-Directed RNA Polymerases antagonists & inhibitors, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Respiratory Syncytial Viruses enzymology, Viral Proteins antagonists & inhibitors
- Abstract
Targeting viral polymerases has been a proven and attractive strategy for antiviral drug discovery. Herein we describe our effort in improving the antiviral activity and physical properties of a series of benzothienoazepine compounds as respiratory syncytial virus (RSV) RNA polymerase inhibitors. The antiviral activity and spectrum of this class was significantly improved by exploring the amino substitution of the pyridine ring, resulting in the discovery of the most potent RSV A polymerase inhibitors reported to date., (Copyright © 2013. Published by Elsevier Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
20. Analysis of SAT1 type foot-and-mouth disease virus capsid proteins: influence of receptor usage on the properties of virus particles.
- Author
-
Maree FF, Blignaut B, Aschenbrenner L, Burrage T, and Rieder E
- Subjects
- Amino Acid Sequence, Amino Acid Substitution, Animals, CHO Cells, Capsid Proteins metabolism, Cricetinae, Cricetulus, Foot-and-Mouth Disease Virus immunology, Molecular Sequence Data, Mutation, Protein Structure, Tertiary, Sequence Alignment, Virion ultrastructure, Capsid Proteins chemistry, Capsid Proteins genetics, Foot-and-Mouth Disease Virus genetics, Foot-and-Mouth Disease Virus metabolism, Receptors, Virus metabolism
- Abstract
The three SAT serotype viruses, endemic in Africa, are well known for their difficulty to adapt to cell culture. The viral mechanism involved in foot-and-mouth disease virus (FMDV) tissue tropism and cell-entry is not well understood. A recombinant, small plaque-forming virus (vSAT1tc), derived from a tissue culture-adapted SAT1 virus (SAR/9/81tc), revealed four amino acid substitutions (VP3 Asp192→Tyr; VP3 Ser217→Ile; VP1 Ala69→Gly and VP1 Asn110→Lys) in the capsid, compared to the SAR/9/81wt isolate collected from infected impala epithelium. One substitution added a positively charged lysine residue to the short βF-βG loop of VP1. Furthermore, vSAT1tc displayed a high affinity for CHO-K1 cells possibly via interaction with negatively charged sulphated polysaccharides while SAT1 impala strain relied strongly on α(V)β6 integrin receptors for cell entry. The cell culture adaptation and small plaque phenotype of vSAT1tc was accompanied by differences in particle aggregation and significant differences in acid stability. Based on limited cross neutralization data, the antigenic features seem to be unchanged. Thus, acquisition of positively charged residues in the virion may be beneficial for adaptation of SAT type field strains to cell culture., (Copyright © 2010 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
21. A recombinant sialidase fusion protein effectively inhibits human parainfluenza viral infection in vitro and in vivo.
- Author
-
Moscona A, Porotto M, Palmer S, Tai C, Aschenbrenner L, Triana-Baltzer G, Li QX, Wurtman D, Niewiesk S, and Fang F
- Subjects
- Amphiregulin, Animals, Cell Line, Disease Models, Animal, EGF Family of Proteins, Enzyme-Linked Immunosorbent Assay, Glycoproteins genetics, Humans, Intercellular Signaling Peptides and Proteins genetics, Lymphocyte Depletion, Paramyxoviridae Infections enzymology, Phosphoproteins immunology, Rats, Sigmodontinae, Trachea immunology, Trachea virology, Viral Plaque Assay, Viral Proteins immunology, Neuraminidase genetics, Paramyxoviridae Infections immunology, Receptors, Cell Surface genetics, Recombinant Fusion Proteins pharmacology
- Abstract
Background: The first step in infection by human parainfluenza viruses (HPIVs) is binding to the surface of respiratory epithelial cells via interaction between viral receptor-binding molecules and sialic acid-containing receptors. DAS181, a recombinant sialidase protein containing the catalytic domain of Actinomyces viscosus sialidase, removes cell surface sialic acid, and we proposed that it would inhibit HPIV infection., Methods: Depletion of sialic acid receptors by DAS181 was evaluated by lectin-binding assays. Anti-HPIV activity in cultured cell lines and in human airway epithelium was assessed by the reduction in viral genomes and/or plaque forming units on treatment. In vivo efficacy of intranasally administered DAS181 was assessed using a cotton rat model., Results: DAS181-mediated desialylation led to anti-HPIV activity in cell lines and human airway epithelium. Intranasal DAS181 in cotton rats, a model for human disease, significantly curtailed infection., Conclusions: Enzymatic removal of the sialic acid moiety of HPIV receptors inhibits infection with all tested HPIV strains, both in vitro and in cotton rats. Enzyme-mediated removal of sialic acid receptors represents a novel antiviral strategy for HPIV. The results of this study raise the possibility of a broad spectrum antiviral agent for influenza virus and HPIVs.
- Published
- 2010
- Full Text
- View/download PDF
22. Regulation of myosin-VI targeting to endocytic compartments.
- Author
-
Dance AL, Miller M, Seragaki S, Aryal P, White B, Aschenbrenner L, and Hasson T
- Subjects
- Actins metabolism, Adaptor Proteins, Signal Transducing, Alternative Splicing genetics, Animals, Binding Sites, Carrier Proteins metabolism, Cell Line, Chlorocebus aethiops, DNA Primers, DNA, Complementary genetics, Green Fluorescent Proteins, Humans, Immunoblotting, Immunohistochemistry, Microscopy, Fluorescence, Myosin Heavy Chains genetics, Neuropeptides metabolism, Protein Transport physiology, Rats, Swine, Adaptor Proteins, Vesicular Transport metabolism, Alternative Splicing physiology, Myosin Heavy Chains metabolism, Transport Vesicles metabolism
- Abstract
Myosin-VI has been implicated in endocytic trafficking at both the clathrin-coated and uncoated vesicle stages. The identification of alternative splice forms led to the suggestion that splicing defines the vesicle type to which myosin-VI is recruited. In contrast to this hypothesis, we find that in all cell types examined, myosin-VI is associated with uncoated endocytic vesicles, regardless of splice form. GIPC, a PDZ-domain containing adapter protein, co-assembles with myosin-VI on these vesicles. Myosin-VI is only recruited to clathrin-coated vesicles in cells that express high levels of Dab2, a clathrin-binding adapter protein. Overexpression of Dab2 is sufficient to reroute myosin-VI to clathrin-coated pits in cells where myosin-VI is normally associated with uncoated vesicles. In normal rat kidney (NRK) cells, which express high endogenous levels of Dab2, splicing of the globular tail domain further modulates targeting of ectopically expressed myosin-VI. Although myosin-VI can be recruited to clathrin-coated pits, we find no requirement for myosin-VI motor activity in endocytosis in NRK cells. Instead, our data suggest that myosin-VI recruitment to clathrin-coated pits may be an early step in the recruitment of GIPC to the vesicle surface.
- Published
- 2004
- Full Text
- View/download PDF
23. Myosin VI regulates endocytosis of the cystic fibrosis transmembrane conductance regulator.
- Author
-
Swiatecka-Urban A, Boyd C, Coutermarsh B, Karlson KH, Barnaby R, Aschenbrenner L, Langford GM, Hasson T, and Stanton BA
- Subjects
- Adaptor Proteins, Signal Transducing, Adaptor Proteins, Vesicular Transport metabolism, Apoptosis Regulatory Proteins, Base Sequence, Cell Line, Clathrin metabolism, DNA Primers, Genes, Tumor Suppressor, Humans, Protein Binding, Reverse Transcriptase Polymerase Chain Reaction, Trachea cytology, Trachea metabolism, Tumor Suppressor Proteins, Cystic Fibrosis Transmembrane Conductance Regulator physiology, Endocytosis physiology, Myosin Heavy Chains physiology
- Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a cyclic AMP-regulated Cl(-) channel expressed in the apical plasma membrane in fluid-transporting epithelia. Although CFTR is rapidly endocytosed from the apical membrane of polarized epithelial cells and efficiently recycled back to the plasma membrane, little is known about the molecular mechanisms regulating CFTR endocytosis and endocytic recycling. Myosin VI, an actin-dependent, minus-end directed mechanoenzyme, has been implicated in clathrin-mediated endocytosis in epithelial cells. The goal of this study was to determine whether myosin VI regulates CFTR endocytosis. Endogenous, apical membrane CFTR in polarized human airway epithelial cells (Calu-3) formed a complex with myosin VI, the myosin VI adaptor protein Disabled 2 (Dab2), and clathrin. The tail domain of myosin VI, a dominant-negative recombinant fragment, displaced endogenous myosin VI from interacting with Dab2 and CFTR and increased the expression of CFTR in the plasma membrane by reducing CFTR endocytosis. However, the myosin VI tail fragment had no effect on the recycling of endocytosed CFTR or on fluid-phase endocytosis. CFTR endocytosis was decreased by cytochalasin D, an actin-filament depolymerizing agent. Taken together, these data indicate that myosin VI and Dab2 facilitate CFTR endocytosis by a mechanism that requires actin filaments.
- Published
- 2004
- Full Text
- View/download PDF
24. Uncoated endocytic vesicles require the unconventional myosin, Myo6, for rapid transport through actin barriers.
- Author
-
Aschenbrenner L, Naccache SN, and Hasson T
- Subjects
- Actins analysis, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Cell Line, Dimethyl Sulfoxide pharmacology, Endocytosis physiology, Endosomes metabolism, Endosomes physiology, Green Fluorescent Proteins analysis, Humans, Microscopy, Fluorescence, Myosin Heavy Chains analysis, Myosin Heavy Chains metabolism, Protein Transport, Thiazoles pharmacology, Thiazolidines, Transferrin metabolism, Transport Vesicles physiology, Transport Vesicles ultrastructure, Actins metabolism, Myosin Heavy Chains physiology, Transport Vesicles metabolism
- Abstract
After clathrin-mediated endocytosis, clathrin removal yields an uncoated vesicle population primed for fusion with the early endosome. Here we present the first characterization of uncoated vesicles and show that myo6, an unconventional myosin, functions to move these vesicles out of actin-rich regions found in epithelial cells. Time-lapse microscopy revealed that myo6-associated uncoated vesicles were motile and exhibited fusion and stretching events before endosome delivery, processes that were dependent on myo6 motor activity. In the absence of myo6 motor activity, uncoated vesicles remained trapped in the actin mesh, where they exhibited Brownian-like motion. Exit from the actin mesh occurred by a slow diffusion-based mechanism, delaying transferrin trafficking to the early endosome. Expression of a myo6 mutant that bound tightly to F-actin produced immobilized vesicles and blocked trafficking. Depolymerization of the actin cytoskeleton rescued this block and specifically accelerated transferrin delivery to the early endosome without affecting earlier steps in endocytosis. Therefore actin is a physical barrier impeding uncoated vesicle trafficking, and myo6 is recruited to move the vesicles through this barrier for fusion with the early endosome.
- Published
- 2004
- Full Text
- View/download PDF
25. Myo6 facilitates the translocation of endocytic vesicles from cell peripheries.
- Author
-
Aschenbrenner L, Lee T, and Hasson T
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, COS Cells, Carrier Proteins metabolism, Cells, Cultured, Chlorocebus aethiops, Cloning, Molecular, Green Fluorescent Proteins, Humans, Luminescent Proteins physiology, Mice, Microscopy, Fluorescence, Myosin Heavy Chains physiology, NIH 3T3 Cells, Neuropeptides metabolism, Protein Binding physiology, Protein Transport physiology, Receptors, Transferrin metabolism, Receptors, Transferrin physiology, Transport Vesicles physiology, rab5 GTP-Binding Proteins metabolism, rab5 GTP-Binding Proteins physiology, Carrier Proteins physiology, Myosin Heavy Chains metabolism, Neuropeptides physiology, Transport Vesicles metabolism
- Abstract
Immunolocalization studies in epithelial cells revealed myo6 was associated with peripherally located vesicles that contained the transferrin receptor. Pulse-chase experiments after transferrin uptake showed that these vesicles were newly uncoated endocytic vesicles and that myo6 was recruited to these vesicles immediately after uncoating. GIPC, a putative myo6 tail binding protein, was also present. Myo6 was not present on early endosomes, suggesting that myo6 has a transient association with endocytic vesicles and is released upon early endosome fusion. Green fluorescent protein (GFP) fused to myo6 as well as the cargo-binding tail (M6tail) alone targeted to the nascent endocytic vesicles. Overexpression of GFP-M6tail had no effect on a variety of organelle markers; however, GFP-M6tail displaced the endogenous myo6 from nascent vesicles and resulted in a significant delay in transferrin uptake. Pulse-chase experiments revealed that transferrin accumulated in uncoated vesicles within the peripheries of transfected cells and that Rab5 was recruited to the surface of these vesicles. Given sufficient time, the transferrin did traffic to the perinuclear sorting endosome. These data suggest that myo6 is an accessory protein required for the efficient transportation of nascent endocytic vesicles from the actin-rich peripheries of epithelial cells, allowing for timely fusion of endocytic vesicles with the early endosome.
- Published
- 2003
- Full Text
- View/download PDF
26. Calcium ion downregulates soluble guanylyl cyclase activity: evidence for a two-metal ion catalytic mechanism.
- Author
-
Serfass L, Carr HS, Aschenbrenner LM, and Burstyn JN
- Subjects
- Binding Sites, Cations, Divalent metabolism, Cations, Divalent pharmacology, Down-Regulation, Enzyme Activation, Gene Expression Regulation, Enzymologic, Kinetics, Magnesium metabolism, Models, Chemical, Nitric Oxide metabolism, Solubility, Calcium pharmacology, Cyclic GMP metabolism, Guanylate Cyclase antagonists & inhibitors
- Abstract
The inhibition of soluble guanylyl cyclase by Ca2+ has been kinetically characterized and the results support a two-metal-ion catalytic mechanism for formation of cGMP. Ca2+ reversibly inhibits both the basal and NO-stimulated forms of bovine lung soluble guanylyl cyclase. Inhibition is independent of the activator identity and concentration, revealing that Ca2+ interacts with a site independent of the heme regulatory site. Inhibition by Ca2+ is competitive with respect to Mg2+ in excess of substrate, with Kis values of 29 +/- 4 and 6.6 +/- 0.6 microM for the basal and activated states, respectively. Ca2+ inhibits noncompetitively with respect to the substrate MgGTP in both activity states. The qualitatively similar inhibition pattern and quantitatively different Ki values between the basal and NO-stimulated states suggest that the Ca2+ binding site undergoes some structural modification upon activation of the enzyme. The competitive nature of Ca2+ inhibition with respect to excess Mg2+ is consistent with a two-metal-ion mechanism for cyclization.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.