1
|
Shih TM, Munoz C, Acon-Chen C, Keith ZM. Pharmacology of Adenosine A 1 Receptor Agonist in a Humanized Esterase Mouse Seizure Model Following Soman Intoxication. Neurotox Res 2024; 42:41. [PMID: 39230655 PMCID: PMC11374867 DOI: 10.1007/s12640-024-00717-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 06/14/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024]
Abstract
Recently a novel genetically modified mouse strain with serum carboxylesterase knocked-out and the human acetylcholinesterase gene knocked-in (KIKO) was created to simulate human responses to nerve agent (NA) exposure and its standard medical treatment. A1 adenosine receptor (A1AR) agonist N-bicyclo-(2.2.1)-hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA) alone is a potent anticonvulsant and neuroprotectant (A/N) in both rat and KIKO mouse soman (GD) seizure models. In this study we utilized the KIKO mouse to evaluate further the basic pharmacologic A/N effects of ENBA as an adjunct to standard NA medical treatments (i.e., atropine sulfate, pralidoxime chloride [2-PAM], and midazolam). Male mice, implanted with cortical electroencephalographic (EEG) electrodes, were pretreated with asoxime (HI-6) and exposed to an epileptogenic dose of GD (33 µg/kg, s.c.) or saline (sham exposure) and then treated 15 min after seizure onset with ENBA at 15 mg/kg, i.p. (a minimum efficacy dose in suppressing NA-induced seizure) alone or as an adjunct to standard medical treatments. We collected EEG activity, seizure suppression outcomes, daily body temperature and weight, heart rate, toxic signs, neuropathology, and lethality data for up to 14 days. Without ENBA, death from NA exposure was 45%, while with ENBA, either alone or in combination with midazolam, the survival improved to 80% and 90%, respectively. Additionally, seizure was suppressed quickly and permanently, toxic signs, hypothermia, and bradycardia recovered by 48 h, and no neuropathology was evident. Our findings confirmed that ENBA is a potent A/N adjunct for delayed medical treatments of NA exposure.
Collapse
Affiliation(s)
- Tsung-Ming Shih
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Crystal Munoz
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA
- University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9162, USA
| | - Cindy Acon-Chen
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA
| | - Zora-Maya Keith
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, 21010-5400, USA
| |
Collapse
|
2
|
Harkins J, Langston J, Keith ZM, Munoz C, Acon-Chen C, Shih TM. Learning and memory function preserved by delayed A 1 adenosine receptor agonist treatment following soman intoxication in rats and a humanized esterase mouse model. Neuropharmacology 2024; 253:109983. [PMID: 38704023 PMCID: PMC11132435 DOI: 10.1016/j.neuropharm.2024.109983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
Exposure to organophosphorus compounds, such as soman (GD), cause widespread toxic effects, sustained status epilepticus, neuropathology, and death. The A1 adenosine receptor agonist N-bicyclo-(2.2.1)-hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA), when given 1 min after GD exposure, provides neuroprotection and prevents behavioral impairments. Here, we tested the ability of ENBA at delayed treatment times to improve behavioral outcomes via a two-way active avoidance task in two male animal models, each consisting of saline and GD exposure groups. In a rat model, animals received medical treatments (atropine sulfate [A], 2-PAM [P], and midazolam [MDZ]) or AP + MDZ + ENBA at 15 or 30 min after seizure onset and were subjected to behavioral testing for up to 14 days. In a human acetylcholinesterase knock-in serum carboxylesterase knock-out mouse model, animals received AP, AP + MDZ, AP + ENBA, or AP + MDZ + ENBA at 15 min post seizure onset and were subjected to the behavioral task on days 7 and 14. In rats, the GD/AP + MDZ + ENBA group recovered to saline-exposed avoidance levels while the GD/AP + MDZ group did not. In mice, in comparison with GD/AP + MDZ group, the GD/AP + MDZ + ENBA showed decreases in escape latency, response latency, and pre-session crossings, as well as increases in avoidances. In both models, only ENBA-treated groups showed control level inter-trial interval crossings by day 14. Our findings suggest that ENBA, alone and as an adjunct to medical treatments, can improve behavioral and cognitive outcomes when given at delayed time points after GD intoxication.
Collapse
Affiliation(s)
- Joshua Harkins
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Jeffrey Langston
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Zora-Maya Keith
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Crystal Munoz
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Cindy Acon-Chen
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| | - Tsung-Ming Shih
- Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD, 21010-5400, USA.
| |
Collapse
|
3
|
Wang H, Qiu J, Zhou M, Luo Y, Li X, Wang M. Monobutyrin Can Regulate the Gut Microbiota, Which Is Beneficial for the Development of Intestinal Barrier Function and Intestinal Health in Weaned Mice. Nutrients 2024; 16:2052. [PMID: 38999800 PMCID: PMC11243092 DOI: 10.3390/nu16132052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/14/2024] Open
Abstract
In this study, we investigated the effect of monobutyrin (MB) on the gut microbiota and intestinal health of weaned mice. MB was administered via gavage to 21-day-old weaned mice. Samples of small intestinal and ileal contents were collected on day 1, day 7, and day 21 post-administration. Seven days of MB administration enhanced the mucin layer and morphological structure of the intestine and the integrity of the intestinal brush border. Both MB and sodium butyrate (SB) accelerated tight junction development. Compared to SB, MB modulated intestinal T cells in a distinct manner. MB increased the ratio of Treg cells in the small intestine upon the cessation of weaning. After 21 days of MB administration, enhancement of the villus structure of the ileum was observed. MB increased the proportion of Th17 cells in the ileum. MB facilitated the transition of the small intestinal microbiota toward an adult microbial community structure and enhanced the complexity of the microbial community structure. An increase in Th17 cells enhanced intestinal barrier function. The regulatory effect of MB on Th17 cells may occur through the intestinal microbiota. Therefore, MB can potentially be used to promote intestinal barrier function, especially for weaning animals, with promising application prospects.
Collapse
Affiliation(s)
- Haidong Wang
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ji Qiu
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Minyao Zhou
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanqiu Luo
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyu Li
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Minqi Wang
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
4
|
Nagaoka M, Sakai Y, Nakajima M, Fukami T. Role of carboxylesterase and arylacetamide deacetylase in drug metabolism, physiology, and pathology. Biochem Pharmacol 2024; 223:116128. [PMID: 38492781 DOI: 10.1016/j.bcp.2024.116128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/20/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
Carboxylesterases (CES1 and CES2) and arylacetamide deacetylase (AADAC), which are expressed primarily in the liver and/or gastrointestinal tract, hydrolyze drugs containing ester and amide bonds in their chemical structure. These enzymes often catalyze the conversion of prodrugs, including the COVID-19 drugs remdesivir and molnupiravir, to their pharmacologically active forms. Information on the substrate specificity and inhibitory properties of these enzymes, which would be useful for drug development and toxicity avoidance, has accumulated. Recently,in vitroandin vivostudies have shown that these enzymes are involved not only in drug hydrolysis but also in lipid metabolism. CES1 and CES2 are capable of hydrolyzing triacylglycerol, and the deletion of their orthologous genes in mice has been associated with impaired lipid metabolism and hepatic steatosis. Adeno-associated virus-mediated human CES overexpression decreases hepatic triacylglycerol levels and increases fatty acid oxidation in mice. It has also been shown that overexpression of CES enzymes or AADAC in cultured cells suppresses the intracellular accumulation of triacylglycerol. Recent reports indicate that AADAC can be up- or downregulated in tumors of various organs, and its varied expression is associated with poor prognosis in patients with cancer. Thus, CES and AADAC not only determine drug efficacy and toxicity but are also involved in pathophysiology. This review summarizes recent findings on the roles of CES and AADAC in drug metabolism, physiology, and pathology.
Collapse
Affiliation(s)
- Mai Nagaoka
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshiyuki Sakai
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
5
|
Zhang W, Oh JH, Zhang W, Aldrich CC, Sirianni RW, Elmquist WF. Pharmacokinetics of panobinostat: Inter-species difference in metabolic stability. J Pharmacol Exp Ther 2024; 389:JPET-AR-2023-002051. [PMID: 38409112 PMCID: PMC10949161 DOI: 10.1124/jpet.123.002051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/13/2024] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
Panobinostat is a potent pan-HDAC inhibitor that has been tested in multiple studies for the treatment of brain tumors. There have been contrasting views surrounding its efficacy for the treatment of tumors in the CNS following systemic administration when examined in different models or species. We conducted experiments using three different mouse strains or genotypes to have a more comprehensive understanding of the systemic as well as the CNS distributional kinetics of panobinostat. Our study found that panobinostat experienced rapid degradation in vitro in FVB mouse matrices and a faster degradation rate was observed at 37{degree sign}C compared with room temperature and 4{degree sign}C, suggesting that the in vitro instability of panobinostat was due to enzymatic metabolism. Panobinostat also showed inter-strain and inter-species differences in the in vitro plasma stability; and was stable in human plasma. The objective of this study was to examine the in vitro metabolic stability of panobinostat in different matrices and assess the influence of that metabolic stability on the in vivo pharmacokinetics and CNS delivery of panobinostat. Importantly, the plasma stability in various mouse strains was not reflected in the in vivo systemic pharmacokinetic behavior of panobinostat. Several hypotheses arise from this finding, including: the binding of panobinostat to red blood cells, the existence of competing endogenous compounds to enzyme(s), the distribution into tissues with a lower level of enzymatic activity or the metabolism occurring in the plasma is a small fraction of the total metabolism in vivo Significance Statement Panobinostat showed different in vitro degradation in plasma from different mouse strains and genotypes. However, despite the differences surrounding in vitro plasma stability, panobinostat showed similar in vivo pharmacokinetic behavior in different mouse models. This suggests that the inter-strain difference in enzymatic activity did not affect the in vivo pharmacokinetic behavior of panobinostat and its CNS distribution in mice. This lack of translation between in vitro metabolism assays and in vivo disposition can confound drug development.
Collapse
Affiliation(s)
- Wenqiu Zhang
- Pharmaceutics, University of Minnesota, United States
| | - Ju-Hee Oh
- University of Minnesota, United States
| | | | | | | | | |
Collapse
|
6
|
Smolak P, Nguyen M, Diamond C, Wescott H, Doedens JR, Schooley K, Snouwaert JN, Bock MG, Harrison D, Watt AP, Koller BH, Gabel CA. Target Cell Activation of a Structurally Novel NOD-Like Receptor Pyrin Domain-Containing Protein 3 Inhibitor NT-0796 Enhances Potency. J Pharmacol Exp Ther 2024; 388:798-812. [PMID: 38253384 DOI: 10.1124/jpet.123.001941] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Abstract
The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome is a central regulator of innate immunity, essential for processing and release of interleukin-1β and pyroptotic cell death. As endogenous NLRP3 activating triggers are hallmarks of many human chronic inflammatory diseases, inhibition of NLRP3 has emerged as a therapeutic target. Here we identify NDT-19795 as a novel carboxylic acid-containing NLRP3 activation inhibitor in both human and mouse monocytes and macrophages. Remarkably, conversion of the carboxylate to an isopropyl-ester (NT-0796) greatly enhances NLRP3 inhibitory potency in human monocytes. This increase is attributed to the ester-containing pharmacophore being more cell-penetrant than the acid species and, once internalized, the ester being metabolized to NDT-19795 by carboxylesterase-1 (CES-1). Mouse macrophages do not express CES-1, and NT-0796 is ineffective in these cells. Mice also contain plasma esterase (Ces1c) activity which is absent in humans. To create a more human-like model, we generated a mouse line in which the genome was modified, removing Ces1c and replacing this segment of DNA with the human CES-1 gene driven by a mononuclear phagocyte-specific promoter. We show human CES-1 presence in monocytes/macrophages increases the ability of NT-0796 to inhibit NLRP3 activation both in vitro and in vivo. As NLRP3 is widely expressed by monocytes/macrophages, the co-existence of CES-1 in these same cells affords a unique opportunity to direct ester-containing NLRP3 inhibitors precisely to target cells of interest. Profiling NT-0796 in mice humanized with respect to CES-1 biology enables critical modeling of the pharmacokinetics and pharmacodynamics of this novel therapeutic candidate. SIGNIFICANCE STATEMENT: Inhibition of NLRP3 represents a desirable therapeutic strategy for the treatment of multiple human disorders. In this study pharmacological properties of a structurally-novel, ester-containing NLRP3 inhibitor NT-0796 are characterized. To study pharmacodynamics of NT-0796 in vivo, a mouse line was engineered possessing more human-like traits with respect to carboxylesterase biology. In the context of these hCES-1 mice, NT-0796 serves as a more effective inhibitor of NLRP3 activation than the corresponding acid, highlighting the full translational potential of the ester strategy.
Collapse
Affiliation(s)
- Pamela Smolak
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - MyTrang Nguyen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - Christine Diamond
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - Heather Wescott
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - John R Doedens
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - Kenneth Schooley
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - John N Snouwaert
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - Mark G Bock
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - David Harrison
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - Alan P Watt
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - Beverly H Koller
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| | - Christopher A Gabel
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (B.H.K., M.N., J.N.S.); Nodthera, Seattle Washington (P.S., C.D., H.W., J.R.D., K.S., C.A.G.); Nodthera, Cambridge, United Kingdom (D.H., A.P.W.); and Nodthera, Boston, Massachusetts (M.G.B.)
| |
Collapse
|
7
|
Feng Y, Lee J, Yang L, Hilton MB, Morris K, Seaman S, Edupuganti VVSR, Hsu KS, Dower C, Yu G, So D, Bajgain P, Zhu Z, Dimitrov DS, Patel NL, Robinson CM, Difilippantonio S, Dyba M, Corbel A, Basuli F, Swenson RE, Kalen JD, Suthe SR, Hussain M, Italia JS, Souders CA, Gao L, Schnermann MJ, St Croix B. Engineering CD276/B7-H3-targeted antibody-drug conjugates with enhanced cancer-eradicating capability. Cell Rep 2023; 42:113503. [PMID: 38019654 PMCID: PMC10872261 DOI: 10.1016/j.celrep.2023.113503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/18/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
CD276/B7-H3 represents a promising target for cancer therapy based on widespread overexpression in both cancer cells and tumor-associated stroma. In previous preclinical studies, CD276 antibody-drug conjugates (ADCs) exploiting a talirine-type pyrrolobenzodiazepine (PBD) payload showed potent activity against various solid tumors but with a narrow therapeutic index and dosing regimen higher than that tolerated in clinical trials using other antibody-talirine conjugates. Here, we describe the development of a modified talirine PBD-based fully human CD276 ADC, called m276-SL-PBD, that is cross-species (human/mouse) reactive and can eradicate large 500-1,000-mm3 triple-negative breast cancer xenografts at doses 10- to 40-fold lower than the maximum tolerated dose. By combining CD276 targeting with judicious genetic and chemical ADC engineering, improved ADC purification, and payload sensitivity screening, these studies demonstrate that the therapeutic index of ADCs can be substantially increased, providing an advanced ADC development platform for potent and selective targeting of multiple solid tumor types.
Collapse
Affiliation(s)
- Yang Feng
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Jaewon Lee
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Liping Yang
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Mary Beth Hilton
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA; Basic Research Program, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Karen Morris
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA; Basic Research Program, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Steven Seaman
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | | | - Kuo-Sheng Hsu
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Christopher Dower
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Guojun Yu
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Daeho So
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Pradip Bajgain
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Zhongyu Zhu
- Protein Interactions Section, Cancer and Inflammation Program, NCI, NIH, Frederick, MD 21702, USA
| | - Dimiter S Dimitrov
- Protein Interactions Section, Cancer and Inflammation Program, NCI, NIH, Frederick, MD 21702, USA
| | - Nimit L Patel
- Small Animal Imaging Program, FNLCR, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Christina M Robinson
- Animal Research Technical Support, FNLCR, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Simone Difilippantonio
- Animal Research Technical Support, FNLCR, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Marzena Dyba
- Biophysics Resource in the Center for Structural Biology, NCI, NIH, Frederick, MD, USA
| | - Amanda Corbel
- Invention Development Program, Technology Transfer Center, NCI, Frederick, MD 21701, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, NIH, Rockville, MD 20850, USA
| | - Rolf E Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, NIH, Rockville, MD 20850, USA
| | - Joseph D Kalen
- Small Animal Imaging Program, FNLCR, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | | | | | | | | | - Ling Gao
- Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA
| | - Martin J Schnermann
- Organic Synthesis Section, Chemical Biology Laboratory, CCR, NCI, Frederick, MD 21702, USA
| | - Brad St Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA.
| |
Collapse
|
8
|
Masson P, Shaihutdinova Z, Lockridge O. Drug and pro-drug substrates and pseudo-substrates of human butyrylcholinesterase. Biochem Pharmacol 2023; 218:115910. [PMID: 37972875 DOI: 10.1016/j.bcp.2023.115910] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Butyrylcholinesterase (BChE) is present in plasma and numerous cells and organs. Its physiological function(s) is(are) still unclear. However, this enzyme is of pharmacological and toxicological importance. It displays a broad specificity and is capable of hydrolyzing a wide range of substrates with turnovers differing by several orders of magnitude. Nowaday, these substrates include more than two dozen carboxyl-ester drugs, numerous acetylated prodrugs, and transition state analogues of acetylcholine. In addition, BChE displays a promiscuous hydrolytic activity toward amide bonds of arylacylamides, and slowly hydrolyzes carbamyl- and phosphoryl-esters. Certain pseudo-substrates like carbamates and organophosphates are major drugs of potential medical interest. The existence of a large genetic poly-allelism, affecting the catalytic properties of BChE is at the origin of clinical complications in the use of certain drugs catabolized by BChE. The number of drugs and prodrugs hydrolyzed by BChE is expected to increase in the future. However, very few quantitative data (Km, kcat) are available for most marketed drugs, and except for myorelaxants like succinylcholine and mivacurium, the impact of BChE genetic mutations on catalytic parameters has not been evaluated for most of these drugs.
Collapse
Affiliation(s)
- Patrick Masson
- Laboratory of Biochemical Neuropharmacology, Kazan Federal University, Kazan, Russian Federation.
| | - Zukhra Shaihutdinova
- Laboratory of Biochemical Neuropharmacology, Kazan Federal University, Kazan, Russian Federation
| | - Oksana Lockridge
- Eppley Institute, University of Nebraska Medical Center, Omaha NE, USA
| |
Collapse
|
9
|
Novotná K, Tenora L, Prchalová E, Paule J, Alt J, Veeravalli V, Lam J, Wu Y, Šnajdr I, Gori S, Mettu VS, Tsukamoto T, Majer P, Slusher BS, Rais R. Discovery of tert-Butyl Ester Based 6-Diazo-5-oxo-l-norleucine Prodrugs for Enhanced Metabolic Stability and Tumor Delivery. J Med Chem 2023; 66:15493-15510. [PMID: 37949450 PMCID: PMC10683027 DOI: 10.1021/acs.jmedchem.3c01681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
The glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON) exhibits remarkable anticancer efficacy; however, its therapeutic potential is hindered by its toxicity to gastrointestinal (GI) tissues. We recently reported the discovery of DRP-104, a tumor-targeted DON prodrug with excellent efficacy and tolerability, which is currently in clinical trials. However, DRP-104 exhibits limited aqueous solubility, and the instability of its isopropyl ester promoiety leads to the formation of an inactive M1-metabolite, reducing overall systemic prodrug exposure. Herein, we aimed to synthesize DON prodrugs with various ester and amide promoieties with improved solubility, GI stability, and DON tumor delivery. Twenty-one prodrugs were synthesized and characterized in stability and pharmacokinetics studies. Of these, P11, tert-butyl-(S)-6-diazo-2-((S)-2-(2-(dimethylamino)acetamido)-3-phenylpropanamido)-5-oxo-hexanoate, showed excellent metabolic stability in plasma and intestinal homogenate, high aqueous solubility, and high tumor DON exposures and preserved the ideal tumor-targeting profile of DRP-104. In conclusion, we report a new generation of glutamine antagonist prodrugs with improved physicochemical and pharmacokinetic attributes.
Collapse
Affiliation(s)
- Kateřina Novotná
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Institute
of Organic Chemistry and Biochemistry v.v.i., Academy of Sciences
of the Czech Republic, Prague 160 00, Czech Republic
- Department
of Organic Chemistry, Faculty of Science, Charles University, Prague 128 00, Czech Republic
| | - Lukáš Tenora
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Institute
of Organic Chemistry and Biochemistry v.v.i., Academy of Sciences
of the Czech Republic, Prague 160 00, Czech Republic
| | - Eva Prchalová
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Institute
of Organic Chemistry and Biochemistry v.v.i., Academy of Sciences
of the Czech Republic, Prague 160 00, Czech Republic
| | - James Paule
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Jesse Alt
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Vijay Veeravalli
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Jenny Lam
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Ying Wu
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Ivan Šnajdr
- Institute
of Organic Chemistry and Biochemistry v.v.i., Academy of Sciences
of the Czech Republic, Prague 160 00, Czech Republic
| | - Sadakatali Gori
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Vijaya Saradhi Mettu
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Takashi Tsukamoto
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Pavel Majer
- Institute
of Organic Chemistry and Biochemistry v.v.i., Academy of Sciences
of the Czech Republic, Prague 160 00, Czech Republic
| | - Barbara S. Slusher
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Rana Rais
- Johns
Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Pharmacology and
Molecular Sciences, Neuroscience, Medicine, and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
10
|
Niquet J, Nguyen D, de Araujo Furtado M, Lumley L. Treatment of cholinergic-induced status epilepticus with polytherapy targeting GABA and glutamate receptors. Epilepsia Open 2023; 8 Suppl 1:S117-S140. [PMID: 36807554 PMCID: PMC10173853 DOI: 10.1002/epi4.12713] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
Despite new antiseizure medications, the development of cholinergic-induced refractory status epilepticus (RSE) continues to be a therapeutic challenge as pharmacoresistance to benzodiazepines and other antiseizure medications quickly develops. Studies conducted by Epilepsia. 2005;46:142 demonstrated that the initiation and maintenance of cholinergic-induced RSE are associated with trafficking and inactivation of gamma-aminobutyric acid A receptors (GABAA R) thought to contribute to the development of benzodiazepine pharmacoresistance. In addition, Dr. Wasterlain's laboratory reported that increased N-methyl-d-aspartate receptors (NMDAR) and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR) contribute to enhanced glutamatergic excitation (Neurobiol Dis. 2013;54:225; Epilepsia. 2013;54:78). Thus, Dr. Wasterlain postulated that targeting both maladaptive responses of reduced inhibition and increased excitation that is associated with cholinergic-induced RSE should improve therapeutic outcome. We currently review studies in several animal models of cholinergic-induced RSE that demonstrate that benzodiazepine monotherapy has reduced efficacy when treatment is delayed and that polytherapy with drugs that include a benzodiazepine (eg midazolam and diazepam) to counter loss of inhibition, concurrent with an NMDA antagonist (eg ketamine) to reduce excitation provide improved efficacy. Improved efficacy with polytherapy against cholinergic-induced seizure is demonstrated by reduction in (1) seizure severity, (2) epileptogenesis, and (3) neurodegeneration compared with monotherapy. Animal models reviewed include pilocarpine-induced seizure in rats, organophosphorus nerve agent (OPNA)-induced seizure in rats, and OPNA-induced seizure in two mouse models: (1) carboxylesterase knockout (Es1-/- ) mice which, similarly to humans, lack plasma carboxylesterase and (2) human acetylcholinesterase knock-in carboxylesterase knockout (KIKO) mice. We also review studies showing that supplementing midazolam and ketamine with a third antiseizure medication (valproate or phenobarbital) that targets a nonbenzodiazepine site rapidly terminates RSE and provides further protection against cholinergic-induced SE. Finally, we review studies on the benefits of simultaneous compared with sequential drug treatments and the clinical implications that lead us to predict improved efficacy of early combination drug therapies. The data generated from seminal rodent studies of efficacious treatment of cholinergic-induced RSE conducted under Dr. Wasterlain's guidance suggest that future clinical trials should treat the inadequate inhibition and temper the excess excitation that characterize RSE and that early combination therapies may provide improved outcome over benzodiazepine monotherapy.
Collapse
Affiliation(s)
- Jerome Niquet
- Department of NeurologyDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
- Epilepsy Research LaboratoryVeterans Affairs Greater Los Angeles Healthcare SystemLos AngelesCaliforniaUSA
| | - Donna Nguyen
- Neuroscience DepartmentU.S. Army Medical Research Institute of Chemical Defense (USAMRICD)Aberdeen Proving GroundMarylandUSA
| | | | - Lucille Lumley
- Neuroscience DepartmentU.S. Army Medical Research Institute of Chemical Defense (USAMRICD)Aberdeen Proving GroundMarylandUSA
| |
Collapse
|
11
|
Shoaib M, Kim N, Choudhary RC, Espin B, Nishikimi M, Iverson A, Yagi T, Marashi Shoshtari SS, Shinozaki K, Becker LB, Kim J. Continuously increased generation of ROS in human plasma after cardiac arrest as determined by Amplex Red oxidation. Free Radic Res 2023; 57:384-394. [PMID: 37642450 DOI: 10.1080/10715762.2023.2250547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/23/2023] [Accepted: 08/13/2023] [Indexed: 08/31/2023]
Abstract
Oxidative stress is believed to be a major cause of injury after cardiac arrest (CA). While the effects of ROS generated within tissues have been extensively investigated, the potential of plasma-generated ROS in contributing to CA pathology has not been examined. We utilized Amplex Red (AR) to measure the real time-generation of ROS in isolated plasma from human CA patients. We first used post-CA rat plasma to identify interfering factors for AR oxidation, and then applied this knowledge to analyze human plasma samples, accounting for the identified confounders. We found significantly increased AR oxidation rates lasting for 4 h in post-CA rat plasma compared to baseline. AR oxidation was unchanged with removal of horseradish peroxidase or addition of catalase. However, adding carboxylesterase inhibitors significantly decreased AR oxidation in rat plasma, which implicated increased carboxylesterase activity, not ROS leading to increased AR oxidation. AR oxidation rates were also significantly increased in human CA patient plasma compared to control and this increase persisted even with carboxylesterase inhibition, suggesting continuously increased ROS-generation within plasma post-CA in humans. The increased ROS generation may be one major source of injury post-CA that may be mitigated with antioxidative therapeutic strategies that can manage the ROS systemically generated in plasma over time.KEY POLICY HIGHLIGHTSWe examined the potential of plasma as a source of ROS generation post-cardiac arrestRat cardiac arrest was used to guide the application of Amplex Red in human plasmaROS generation in plasma is significantly increased after cardiac arrest in humansScavenging excessive ROS in post-resuscitation plasma may improve outcomes of patients.
Collapse
Affiliation(s)
- Muhammad Shoaib
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Nancy Kim
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Rishabh C Choudhary
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Blanca Espin
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Mitsuaki Nishikimi
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Ann Iverson
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Tsukasa Yagi
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | - Koichiro Shinozaki
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Department of Emergency Medicine, Northwell Health, NY, USA
| | - Lance B Becker
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Department of Emergency Medicine, Northwell Health, NY, USA
| | - Junhwan Kim
- Laboratory for Critical Care Physiology, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Department of Emergency Medicine, Northwell Health, NY, USA
| |
Collapse
|
12
|
Shih TM. A novel genetically modified mouse seizure model for evaluating anticonvulsive and neuroprotective efficacy of an A 1 adenosine receptor agonist following soman intoxication. Toxicol Appl Pharmacol 2023; 464:116437. [PMID: 36849019 PMCID: PMC10228141 DOI: 10.1016/j.taap.2023.116437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 01/27/2023] [Accepted: 02/19/2023] [Indexed: 02/27/2023]
Abstract
Recently a novel humanized mouse strain has been successfully generated, in which serum carboxylesterase (CES) knock out (KO) mice (Es1-/-) were further genetically modified by knocking in (KI), or adding, the gene that encodes the human form of acetylcholinesterase (AChE). The resulting human AChE KI and serum CES KO (or KIKO) mouse strain should not only exhibit organophosphorus nerve agent (NA) intoxication in a manner more similar to humans, but also display AChE-specific treatment responses more closely mimicking those of humans to facilitate data translation to pre-clinic trials. In this study, we utilized the KIKO mouse to develop a seizure model for NA medical countermeasure investigation, and then applied it to evaluate the anticonvulsant and neuroprotectant (A/N) efficacy of a specific A1 adenosine receptor (A1AR) agonist, N-bicyclo-(2.2.1)hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA), which has been shown in a rat seizure model to be a potent A/N compound. Male mice surgically implanted with cortical electroencephalographic (EEG) electrodes a week earlier were pretreated with HI-6 and challenged with various doses (26 to 47 μg/kg, SC) of soman (GD) to determine a minimum effective dose (MED) that induced sustained status epilepticus (SSE) activity in 100% of animals while causing minimum lethality at 24 h. The GD dose selected was then used to investigate the MED doses of ENBA when given either immediately following SSE initiation (similar to wartime military first aid application) or at 15 min after ongoing SSE seizure activity (applicable to civilian chemical attack emergency triage). The selected GD dose of 33 μg/kg (1.4 x LD50) generated SSE in 100% of KIKO mice and produced only 30% mortality. ENBA at a dose as little as 10 mg/kg, IP, caused isoelectric EEG activity within minutes after administration in naïve un-exposed KIKO mice. The MED doses of ENBA to terminate GD-induced SSE activity were determined to be 10 and 15 mg/kg when treatment was given at the time of SSE onset and when seizure activity was ongoing for 15 min, respectively. These doses were much lower than in the non-genetically modified rat model, which required an ENBA dose of 60 mg/kg to terminate SSE in 100% GD-exposed rats. At MED doses, all mice survived for 24 h, and no neuropathology was observed when the SSE was stopped. The findings confirmed that ENBA is a potent A/N for both immediate and delayed (i.e., dual purposed) therapy to victims of NA exposure and serves as a promising neuroprotective antidotal and adjunctive medical countermeasure candidate for pre-clinical research and development for human application.
Collapse
Affiliation(s)
- Tsung-Ming Shih
- Neuroscience Department, Medical Toxicology Research Division, U.S. Army Medical Research Institute of Chemical Defense, Aberdeen, Proving Ground, MD 21010-5400, USA..
| |
Collapse
|
13
|
Ireland D, Rabeler C, Gong T, Collins EMS. Bioactivation and detoxification of organophosphorus pesticides in freshwater planarians shares similarities with humans. Arch Toxicol 2022; 96:3233-3243. [PMID: 36173421 PMCID: PMC10729609 DOI: 10.1007/s00204-022-03387-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/21/2022] [Indexed: 11/02/2022]
Abstract
Organophosphorus pesticides (OPs) are a chemically diverse class of insecticides that inhibit acetylcholinesterase (AChE). Many OPs require bioactivation to their active oxon form via cytochrome P450 to effectively inhibit AChE. OP toxicity can be mitigated by detoxification reactions performed by carboxylesterase and paraoxonase. The relative extent of bioactivation to detoxification varies among individuals and between species, leading to differential susceptibility to OP toxicity. Because of these species differences, it is imperative to characterize OP metabolism in model systems used to assess OP toxicity. We have shown that the asexual freshwater planarian Dugesia japonica is a suitable model to assess OP neurotoxicity and developmental neurotoxicity via rapid, automated testing of adult and developing organisms in parallel using morphological and behavioral endpoints. D. japonica has two cholinesterase enzymes with intermediate properties between AChE and butyrylcholinesterase that are sensitive to OP inhibition. Here, we demonstrate that D. japonica contains the major OP metabolic machinery to be a relevant model for OP neurotoxicity studies. Adult and regenerating D. japonica can bioactivate chlorpyrifos and diazinon into their respective oxons. Significant AChE inhibition was only observed after in vivo metabolic activation but not when the parent OPs were directly added to planarian homogenate using the same concentrations and timing. Using biochemical assays, we found that D. japonica has both carboxylesterase (24 nmol/(min*mg protein)) and paraoxonase (60 pmol/(min*mg protein)) activity. We show that planarian carboxylesterase activity is distinct from cholinesterase activity using benzil and tacrine. These results further support the use of D. japonica for OP toxicity studies.
Collapse
Affiliation(s)
| | | | - TaiXi Gong
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Eva-Maria S Collins
- Department of Biology, Swarthmore College, Swarthmore, PA, USA.
- Department of Physics and Astronomy, Swarthmore College, Swarthmore, PA, USA.
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Center of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Physics, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
14
|
Rais R, Lemberg KM, Tenora L, Arwood ML, Pal A, Alt J, Wu Y, Lam J, Aguilar JMH, Zhao L, Peters DE, Tallon C, Pandey R, Thomas AG, Dash RP, Seiwert T, Majer P, Leone RD, Powell JD, Slusher BS. Discovery of DRP-104, a tumor-targeted metabolic inhibitor prodrug. SCIENCE ADVANCES 2022; 8:eabq5925. [PMID: 36383674 PMCID: PMC9668306 DOI: 10.1126/sciadv.abq5925] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 09/27/2022] [Indexed: 05/23/2023]
Abstract
6-Diazo-5-oxo-l-norleucine (DON) is a glutamine antagonist that suppresses cancer cell metabolism but concurrently enhances the metabolic fitness of tumor CD8+ T cells. DON showed promising efficacy in clinical trials; however, its development was halted by dose-limiting gastrointestinal (GI) toxicities. Given its clinical potential, we designed DON peptide prodrugs and found DRP-104 [isopropyl(S)-2-((S)-2-acetamido-3-(1H-indol-3-yl)-propanamido)-6-diazo-5-oxo-hexanoate] that was preferentially bioactivated to DON in tumor while bioinactivated to an inert metabolite in GI tissues. In drug distribution studies, DRP-104 delivered a prodigious 11-fold greater exposure of DON to tumor versus GI tissues. DRP-104 affected multiple metabolic pathways in tumor, including decreased glutamine flux into the TCA cycle. In efficacy studies, both DRP-104 and DON caused complete tumor regression; however, DRP-104 had a markedly improved tolerability profile. DRP-104's effect was CD8+ T cell dependent and resulted in robust immunologic memory. DRP-104 represents a first-in-class prodrug with differential metabolism in target versus toxicity tissue. DRP-104 is now in clinical trials under the FDA Fast Track designation.
Collapse
Affiliation(s)
- Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kathryn M. Lemberg
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Lukáš Tenora
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic v.v.i., Prague 16000, Czech Republic
| | - Matthew L. Arwood
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Arindom Pal
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Jenny Lam
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | | | - Liang Zhao
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Diane E. Peters
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Carolyn Tallon
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Rajeev Pandey
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ranjeet P. Dash
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Tanguy Seiwert
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic v.v.i., Prague 16000, Czech Republic
| | - Robert D. Leone
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Jonathan D. Powell
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
15
|
Li H, Perino A, Huang Q, Von Alvensleben GVG, Banaei-Esfahani A, Velazquez-Villegas LA, Gariani K, Korbelius M, Bou Sleiman M, Imbach J, Sun Y, Li X, Bachmann A, Goeminne LJE, Gallart-Ayala H, Williams EG, Ivanisevic J, Auwerx J, Schoonjans K. Integrative systems analysis identifies genetic and dietary modulators of bile acid homeostasis. Cell Metab 2022; 34:1594-1610.e4. [PMID: 36099916 PMCID: PMC9534359 DOI: 10.1016/j.cmet.2022.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/22/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022]
Abstract
Bile acids (BAs) are complex and incompletely understood enterohepatic-derived hormones that control whole-body metabolism. Here, we profiled postprandial BAs in the liver, feces, and plasma of 360 chow- or high-fat-diet-fed BXD male mice and demonstrated that both genetics and diet strongly influence BA abundance, composition, and correlation with metabolic traits. Through an integrated systems approach, we mapped hundreds of quantitative trait loci that modulate BAs and identified both known and unknown regulators of BA homeostasis. In particular, we discovered carboxylesterase 1c (Ces1c) as a genetic determinant of plasma tauroursodeoxycholic acid (TUDCA), a BA species with established disease-preventing actions. The association between Ces1c and plasma TUDCA was validated using data from independent mouse cohorts and a Ces1c knockout mouse model. Collectively, our data are a unique resource to dissect the physiological importance of BAs as determinants of metabolic traits, as underscored by the identification of CES1C as a master regulator of plasma TUDCA levels.
Collapse
Affiliation(s)
- Hao Li
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Alessia Perino
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Qingyao Huang
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Giacomo V G Von Alvensleben
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Amir Banaei-Esfahani
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Laura A Velazquez-Villegas
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Karim Gariani
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Melanie Korbelius
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jéromine Imbach
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Yu Sun
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Alexis Bachmann
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Ludger J E Goeminne
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| | - Evan G Williams
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| | - Kristina Schoonjans
- Laboratory of Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
16
|
Pal A, Gori S, Yoo SW, Thomas AG, Wu Y, Friedman J, Tenora L, Bhasin H, Alt J, Haughey N, Slusher BS, Rais R. Discovery of Orally Bioavailable and Brain-Penetrable Prodrugs of the Potent nSMase2 Inhibitor DPTIP. J Med Chem 2022; 65:11111-11125. [PMID: 35930706 PMCID: PMC9980655 DOI: 10.1021/acs.jmedchem.2c00562] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Extracellular vesicles (EVs) can carry pathological cargo and play an active role in disease progression. Neutral sphingomyelinase-2 (nSMase2) is a critical regulator of EV biogenesis, and its inhibition has shown protective effects in multiple disease states. 2,6-Dimethoxy-4-(5-phenyl-4-thiophen-2-yl-1H-imidazol-2-yl)phenol (DPTIP) is one of the most potent (IC50 = 30 nM) inhibitors of nSMase2 discovered to date. However, DPTIP exhibits poor oral pharmacokinetics (PK), limiting its clinical development. To overcome DPTIP's PK limitations, we synthesized a series of prodrugs by masking its phenolic hydroxyl group. When administered orally, the best prodrug (P18) with a 2',6'-diethyl-1,4'-bipiperidinyl promoiety exhibited >fourfold higher plasma (AUC0-t = 1047 pmol·h/mL) and brain exposures (AUC0-t = 247 pmol·h/g) versus DPTIP and a significant enhancement of DPTIP half-life (2 h vs ∼0.5 h). In a mouse model of acute brain injury, DPTIP released from P18 significantly inhibited IL-1β-induced EV release into plasma and attenuated nSMase2 activity. These studies report the discovery of a DPTIP prodrug with potential for clinical translation.
Collapse
Affiliation(s)
- Arindom Pal
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Sadakatali Gori
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Seung-wan Yoo
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Jacob Friedman
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Lukáš Tenora
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Harshit Bhasin
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Norman Haughey
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Oncology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Departments of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Medicine, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Corresponding Authors: . Tel: 410-502-0497. Fax: 410-614-0659 (R.R.), . Tel: 410-614-0662. Fax: 410-614-0659 (B.S.S.)
| | - Rana Rais
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore MD 21205, USA,Corresponding Authors: . Tel: 410-502-0497. Fax: 410-614-0659 (R.R.), . Tel: 410-614-0662. Fax: 410-614-0659 (B.S.S.)
| |
Collapse
|
17
|
Yin C, Sun Z, Ji C, Li F, Wu H. Toxicological effects of tris(1,3-dichloro-2-propyl) phosphate in oyster Crassostrea gigas using proteomic and phosphoproteomic analyses. JOURNAL OF HAZARDOUS MATERIALS 2022; 434:128824. [PMID: 35427976 DOI: 10.1016/j.jhazmat.2022.128824] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/18/2022] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
As a typical organophosphorus pollutant, tris(1,3-dichloro-2-propyl) phosphate (TDCIPP) has been widely detected in aquatic environment. Previous studies showed that protein phosphorylation might be a vital way of TDCIPP to exert multiple toxic effects. However, there is a lack of high-throughput investigations on how TDCIPP affected protein phosphorylation. In this study, the toxicological effects of TDCIPP were explored by proteomic and phosphoproteomic analyses together with traditional means in oysters Crassostrea gigas treated with 0.5, 5 and 50 μg/L TDCIPP for 28 days. Integration of omic analyses revealed that TDCIPP dysregulated transcription, energy metabolism, and apoptosis and cell proliferation by either directly phosphorylating pivotal proteins or phosphorylating their upstream signaling pathways. The U-shaped response of acetylcholinesterase activities suggested the neurotoxicity of TDCIPP in a hormesis manner. What's more, the increase in caspase-9 activity as well as the expression or phosphorylation alterations in eukaryotic translation initiation factor 4E, cell division control protein 42 and transforming growth factor-β1-induced protein indicated the disruption of homeostasis between apoptosis and cell proliferation, which was consistent with the observation of shedding of digestive cells. Overall, combination of proteomic and phosphoproteomic analyses showed the capability of identifying molecular events, which provided new insights into the toxicological mechanisms of TDCIPP.
Collapse
Affiliation(s)
- Chengcheng Yin
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Zuodeng Sun
- Shandong Fisheries Development and Resource Conservation Center, Ji'nan 250013, PR China
| | - Chenglong Ji
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China; Center for Ocean Mega-Science, Chinese Academy of Sciences (CAS), Qingdao 266071, PR China.
| | - Fei Li
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; Center for Ocean Mega-Science, Chinese Academy of Sciences (CAS), Qingdao 266071, PR China
| | - Huifeng Wu
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, PR China; Center for Ocean Mega-Science, Chinese Academy of Sciences (CAS), Qingdao 266071, PR China.
| |
Collapse
|
18
|
Gage M, Rao NS, Samidurai M, Putra M, Vasanthi SS, Meyer C, Wang C, Thippeswamy T. Soman (GD) Rat Model to Mimic Civilian Exposure to Nerve Agent: Mortality, Video-EEG Based Status Epilepticus Severity, Sex Differences, Spontaneously Recurring Seizures, and Brain Pathology. Front Cell Neurosci 2022; 15:798247. [PMID: 35197823 PMCID: PMC8859837 DOI: 10.3389/fncel.2021.798247] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Modeling a real-world scenario of organophosphate nerve agent (OPNA) exposure is challenging. Military personnel are premedicated with pyridostigmine, which led to the development of OPNA models with pyridostigmine/oxime pretreatment to investigate novel therapeutics for acute and chronic effects. However, civilians are not premedicated with pyridostigmine/oxime. Therefore, experimental models without pyridostigmine were developed by other laboratories though often only in males. Following OPNA exposure, prolonged convulsive seizures (CS) or status epilepticus (SE) are concerning. The duration and severity of CS/SE determine the extent of brain injury in survivors even after treating with medical countermeasures (MCM)/antidotes such as atropine, an oxime, and an anticonvulsant such as diazepam/midazolam. In this study, using a large mixed sex cohort of adult male and female rats, without pretreatment, we demonstrate severe SE lasting for >20 min in 82% of the animals in response to soman (GD,132 μg/kg, s.c.). Atropine sulfate (2 mg/kg, i.m.) and HI-6 (125 mg/kg, i.m.) were administered immediately following soman, and midazolam (3 mg/kg, i.m.) 1 h post-exposure. Immediate MCM treatment is impractical in civilian exposure to civilians, but this approach reduces mortality in experimental models. Interestingly, female rats, irrespective of estrous stages, had an average of 44 min CS (stage ≥ 3), while males had an average of 32 min CS during SE, starting from soman exposure to midazolam treatment. However, in telemetry device implanted groups, there were no significant sex differences in SE severity; males had 40 min and females 43 min of continuous CS until midazolam was administered. No animals died prior to midazolam administration and less than 5% died in the first week after soman intoxication. In telemetered animals, there was a direct correlation between EEG changes and behavioral seizures in real-time. In the long-term, convulsive spontaneously recurring seizures (SRS) were observed in 85% of randomly chosen animals. At 4-months post-soman, the brain histology confirmed reactive gliosis and neurodegeneration. The novel findings of this study are that, in non-telemetered animals, the SE severity following soman intoxication was significantly greater in females compared to males and that the estrous cycle did not influence the response.
Collapse
Affiliation(s)
- Meghan Gage
- Neuroscience Interdepartmental Program, Iowa State University, Ames, IA, United States.,Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Nikhil S Rao
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Manikandan Samidurai
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Marson Putra
- Neuroscience Interdepartmental Program, Iowa State University, Ames, IA, United States.,Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Suraj S Vasanthi
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Christina Meyer
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Chong Wang
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Thimmasettappa Thippeswamy
- Neuroscience Interdepartmental Program, Iowa State University, Ames, IA, United States.,Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
19
|
Lin M, Ellis B, Eubanks LM, Janda KD. Pharmacokinetic Approach to Combat the Synthetic Cannabinoid PB-22. ACS Chem Neurosci 2021; 12:2573-2579. [PMID: 34254505 DOI: 10.1021/acschemneuro.1c00360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Synthetic cannabinoids are part of a group of drugs called new psychoactive substances. Most of these cannabinoids are unregulated, and there are no therapeutic treatments for their addictive properties or reversing a potential overdose. Vaccination and catalytic antibodies strategies were investigated to assess their ability to blunt the psychoactive properties of the cannabinoid PB-22. To complement these antibody concentric investigations, we also disclose the discovery of the enzymatic degradation of this cannabinoid. Serum factors including albumin and carboxylesterase were found to catalyze the hydrolysis of PB-22. Affinity, kinetics, animal behavior, and biodistribution studies were utilized to evaluate the efficiency of these pharmacokinetic approaches. Our findings suggest simple antibody binding as the most efficacious means for altering PB-22's effect on the brain. Catalytic approaches only translated to esterases being capable of PB-22's degradation with a catalytic antibody approach providing no proclivity for PB-22's hydrolysis. Pharmacokinetic approaches provide a powerful strategy for treating substance abuse disorders and overdose for drugs where no therapeutic is available.
Collapse
Affiliation(s)
- Mingliang Lin
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, and The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Beverly Ellis
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, and The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Lisa M. Eubanks
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, and The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Kim D. Janda
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, and The Worm Institute for Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
20
|
Zhao S, Wesseling S, Spenkelink B, Rietjens IMCM. Physiologically based kinetic modelling based prediction of in vivo rat and human acetylcholinesterase (AChE) inhibition upon exposure to diazinon. Arch Toxicol 2021; 95:1573-1593. [PMID: 33715020 PMCID: PMC8113213 DOI: 10.1007/s00204-021-03015-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/25/2021] [Indexed: 01/30/2023]
Abstract
The present study predicts in vivo human and rat red blood cell (RBC) acetylcholinesterase (AChE) inhibition upon diazinon (DZN) exposure using physiological based kinetic (PBK) modelling-facilitated reverse dosimetry. Due to the fact that both DZN and its oxon metabolite diazoxon (DZO) can inhibit AChE, a toxic equivalency factor (TEF) was included in the PBK model to combine the effect of DZN and DZO when predicting in vivo AChE inhibition. The PBK models were defined based on kinetic constants derived from in vitro incubations with liver fractions or plasma of rat and human, and were used to translate in vitro concentration-response curves for AChE inhibition obtained in the current study to predicted in vivo dose-response curves. The predicted dose-response curves for rat matched available in vivo data on AChE inhibition, and the benchmark dose lower confidence limits for 10% inhibition (BMDL10 values) were in line with the reported BMDL10 values. Humans were predicted to be 6-fold more sensitive than rats in terms of AChE inhibition, mainly because of inter-species differences in toxicokinetics. It is concluded that the TEF-coded DZN PBK model combined with quantitative in vitro to in vivo extrapolation (QIVIVE) provides an adequate approach to predict RBC AChE inhibition upon acute oral DZN exposure, and can provide an alternative testing strategy for derivation of a point of departure (POD) in risk assessment.
Collapse
Affiliation(s)
- Shensheng Zhao
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| | - Sebastiaan Wesseling
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Bert Spenkelink
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| |
Collapse
|
21
|
Marrero-Rosado BM, Stone MF, de Araujo Furtado M, Schultz CR, Cadieux CL, Lumley LA. Novel Genetically Modified Mouse Model to Assess Soman-Induced Toxicity and Medical Countermeasure Efficacy: Human Acetylcholinesterase Knock-in Serum Carboxylesterase Knockout Mice. Int J Mol Sci 2021; 22:1893. [PMID: 33672922 PMCID: PMC7918218 DOI: 10.3390/ijms22041893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/06/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
The identification of improved medical countermeasures against exposure to chemical warfare nerve agents (CWNAs), a class of organophosphorus compounds, is dependent on the choice of animal model used in preclinical studies. CWNAs bind to acetylcholinesterase and prevent the catalysis of acetylcholine, causing a plethora of peripheral and central physiologic manifestations, including seizure. Rodents are widely used to elucidate the effects of CWNA-induced seizure, albeit with a caveat: they express carboxylesterase activity in plasma. Carboxylesterase, an enzyme involved in the detoxification of some organophosphorus compounds, plays a scavenging role and decreases CWNA availability, thus exerting a protective effect. Furthermore, species-specific amino acid differences in acetylcholinesterase confound studies that use oximes or other compounds to restore its function after inhibition by CWNA. The creation of a human acetylcholinesterase knock-in/serum carboxylesterase knockout (C57BL/6-Ces1ctm1.1LocAChEtm1.1Loc/J; a.k.a KIKO) mouse may facilitate better modeling of CWNA toxicity in a small rodent species. The current studies characterize the effects of exposure to soman, a highly toxic CWNA, and evaluate the efficacy of anti-seizure drugs in this newly developed KIKO mouse model. Data demonstrate that a combination of midazolam and ketamine reduces seizure duration and severity, eliminates the development of spontaneous recurrent seizures, and protects certain brain regions from neuronal damage in a genetically modified model with human relevance to organophosphorus compound toxicity. This new animal model and the results of this study and future studies using it will enhance medical countermeasures development for both defense and homeland security purposes.
Collapse
Affiliation(s)
- Brenda M. Marrero-Rosado
- Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA; (B.M.M.-R.); (M.F.S.); (C.R.S.); (C.L.C.)
| | - Michael F. Stone
- Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA; (B.M.M.-R.); (M.F.S.); (C.R.S.); (C.L.C.)
| | - Marcio de Araujo Furtado
- Anatomy, Physiology and Genetics Department, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA;
- BioSEaD, LLC, Rockville, MD 20850, USA
| | - Caroline R. Schultz
- Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA; (B.M.M.-R.); (M.F.S.); (C.R.S.); (C.L.C.)
| | - C. Linn Cadieux
- Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA; (B.M.M.-R.); (M.F.S.); (C.R.S.); (C.L.C.)
| | - Lucille A. Lumley
- Medical Toxicology Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, USA; (B.M.M.-R.); (M.F.S.); (C.R.S.); (C.L.C.)
| |
Collapse
|
22
|
Kundrick ER, Marrero-Rosado BM, de Araujo Furtado M, Stone M, Schultz CR, Lumley LA. Cannabidiol reduces soman-induced lethality and seizure severity in female plasma carboxylesterase knockout mice treated with midazolam. Neurotoxicology 2020; 82:130-136. [PMID: 33290784 DOI: 10.1016/j.neuro.2020.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/25/2020] [Accepted: 12/02/2020] [Indexed: 11/18/2022]
Abstract
Cannabidiol, approved for treatment of pediatric refractory epilepsy, has anti-seizure effects in various animal seizure models. Chemical warfare nerve agents, including soman, are organophosphorus chemicals that can induce seizure and death if untreated or if treatment is delayed. Our objective was to evaluate whether cannabidiol would ameliorate soman-induced toxicity using a mouse model that similar to humans lacks plasma carboxylesterase. In the present study, adult female plasma carboxylesterase knockout (Es1-/-) mice were pre-treated with cannabidiol (20-150 mg/kg) or vehicle 1 h prior to exposure to a seizure-inducing dose of soman and evaluated for survival and seizure activity. The muscarinic antagonist atropine sulfate and the oxime HI-6 were administered at 1 min after exposure, and the benzodiazepine midazolam was administered at 30 min after seizure onset. Cannabidiol (150 mg/kg) pre-treatment led to a robust increase in survival rate and attenuated body weight loss in soman-exposed mice treated with medical countermeasures, compared to mice pre-treated with vehicle. In addition, mice pretreated with cannabidiol (150 mg/kg) had a modest reduction in seizure severity after midazolam treatment compared to vehicle-pretreated. These findings of improved outcome with cannabidiol administration in a severe seizure model of soman exposure provide additional pre-clinical support for the benefits of cannabidiol against exposure to seizure-inducing chemical agents and suggest cannabidiol may augment the anti-seizure effects of midazolam.
Collapse
Affiliation(s)
- Erica R Kundrick
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, United States
| | - Brenda M Marrero-Rosado
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, United States
| | - Marcio de Araujo Furtado
- Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, United States; BioSEaD, LLC, Rockville, MD, 20850, United States
| | - Michael Stone
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, United States
| | - Caroline R Schultz
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, United States
| | - Lucille A Lumley
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD 21010, United States.
| |
Collapse
|
23
|
Lin YH, Satani N, Hammoudi N, Yan VC, Barekatain Y, Khadka S, Ackroyd JJ, Georgiou DK, Pham CD, Arthur K, Maxwell D, Peng Z, Leonard PG, Czako B, Pisaneschi F, Mandal P, Sun Y, Zielinski R, Pando SC, Wang X, Tran T, Xu Q, Wu Q, Jiang Y, Kang Z, Asara JM, Priebe W, Bornmann W, Marszalek JR, DePinho RA, Muller FL. An enolase inhibitor for the targeted treatment of ENO1-deleted cancers. Nat Metab 2020; 2:1413-1426. [PMID: 33230295 PMCID: PMC7744354 DOI: 10.1038/s42255-020-00313-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 10/15/2020] [Indexed: 12/15/2022]
Abstract
Inhibiting glycolysis remains an aspirational approach for the treatment of cancer. We have previously identified a subset of cancers harbouring homozygous deletion of the glycolytic enzyme enolase (ENO1) that have exceptional sensitivity to inhibition of its redundant paralogue, ENO2, through a therapeutic strategy known as collateral lethality. Here, we show that a small-molecule enolase inhibitor, POMHEX, can selectively kill ENO1-deleted glioma cells at low-nanomolar concentrations and eradicate intracranial orthotopic ENO1-deleted tumours in mice at doses well-tolerated in non-human primates. Our data provide an in vivo proof of principle of the power of collateral lethality in precision oncology and demonstrate the utility of POMHEX for glycolysis inhibition with potential use across a range of therapeutic settings.
Collapse
Affiliation(s)
- Yu-Hsi Lin
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nikunj Satani
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute of Stroke and Cerebrovascular Disease, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Naima Hammoudi
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victoria C Yan
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yasaman Barekatain
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunada Khadka
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey J Ackroyd
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dimitra K Georgiou
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cong-Dat Pham
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenisha Arthur
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Maxwell
- Institutional Analytics & Informatics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Paul G Leonard
- Core for Biomolecular Structure and Function, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Barbara Czako
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Federica Pisaneschi
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pijus Mandal
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuting Sun
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rafal Zielinski
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Susana Castro Pando
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaobo Wang
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Theresa Tran
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Quanyu Xu
- Pharmaceutical Science Facility, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Wu
- Pharmaceutical Science Facility, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yongying Jiang
- Pharmaceutical Science Facility, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhijun Kang
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M Asara
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Waldemar Priebe
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William Bornmann
- Director of Drug Discovery and Development, Advanced Organic Synthesis LLC, Houston, Texas, USA
| | - Joseph R Marszalek
- Center for Co-Clinical Trials, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Florian L Muller
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
24
|
Marrero-Rosado BM, de Araujo Furtado M, Kundrick ER, Walker KA, Stone MF, Schultz CR, Nguyen DA, Lumley LA. Ketamine as adjunct to midazolam treatment following soman-induced status epilepticus reduces seizure severity, epileptogenesis, and brain pathology in plasma carboxylesterase knockout mice. Epilepsy Behav 2020; 111:107229. [PMID: 32575012 PMCID: PMC7541728 DOI: 10.1016/j.yebeh.2020.107229] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/18/2022]
Abstract
Delayed treatment of cholinergic seizure results in benzodiazepine-refractory status epilepticus (SE) that is thought, at least in part, to result from maladaptive trafficking of N-methyl-d-aspartate (NMDA) and gamma-aminobutyric acid type A (GABAA) receptors, the effects of which may be ameliorated by combination therapy with the NMDA receptor antagonist ketamine. Our objective was to establish whether ketamine and midazolam dual therapy would improve outcome over midazolam monotherapy following soman (GD) exposure when evaluated in a mouse model that, similar to humans, lacks plasma carboxylesterase, greatly reducing endogenous scavenging of GD. In the current study, continuous cortical electroencephalographic activity was evaluated in male and female plasma carboxylesterase knockout mice exposed to a seizure-inducing dose of GD and treated with midazolam or with midazolam and ketamine combination at 40 min after seizure onset. Ketamine and midazolam combination reduced GD-induced lethality, seizure severity, and the number of mice that developed spontaneous recurrent seizure (SRS) compared with midazolam monotherapy. In addition, ketamine-midazolam combination treatment reduced GD-induced neuronal degeneration and microgliosis. These results support that combination of antiepileptic drug therapies aimed at correcting the maladaptive GABAA and NMDA receptor trafficking reduces the detrimental effects of GD exposure. Ketamine may be a beneficial adjunct to midazolam in reducing the epileptogenesis and neuroanatomical damage that follows nerve agent exposure and pharmacoresistant SE.
Collapse
Affiliation(s)
- Brenda M. Marrero-Rosado
- U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010
| | - Marcio de Araujo Furtado
- Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814,BioSEaD, LLC. 451 Hungerford Drive, Rockville, MD, 20850
| | - Erica R. Kundrick
- U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010
| | - Katie A. Walker
- U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010
| | - Michael F. Stone
- U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010
| | - Caroline R. Schultz
- U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010
| | - Donna A. Nguyen
- U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010
| | - Lucille A. Lumley
- U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Road, Aberdeen Proving Ground, MD 21010
| |
Collapse
|
25
|
Harrison D, Boutard N, Brzozka K, Bugaj M, Chmielewski S, Cierpich A, Doedens JR, Fabritius CHRY, Gabel CA, Galezowski M, Kowalczyk P, Levenets O, Mroczkowska M, Palica K, Porter RA, Schultz D, Sowinska M, Topolnicki G, Urbanski P, Woyciechowski J, Watt AP. Discovery of a series of ester-substituted NLRP3 inflammasome inhibitors. Bioorg Med Chem Lett 2020; 30:127560. [PMID: 32956781 DOI: 10.1016/j.bmcl.2020.127560] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/25/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
The NLRP3 inflammasome is a component of the innate immune system involved in the production of proinflammatory cytokines. Aberrant activation by a wide range of exogenous and endogenous signals can lead to chronic, low-grade inflammation. It has attracted a great deal of interest as a drug target due to the association with diseases of large unmet medical need such as Alzheimer's disease, Parkinson's disease, arthritis, and cancer. To date, no drugs specifically targeting inhibition of the NLRP3 inflammasome have been approved. In this work, we used the known NLRP3 inflammasome inhibitor CP-456,773 (aka CRID3 or MCC 950) as our starting point and undertook a Structure-Activity Relationship (SAR) analysis and subsequent scaffold-hopping exercise. This resulted in the rational design of a series of novel ester-substituted urea compounds that are highly potent and selective NLRP3 inflammasome inhibitors, as exemplified by compounds 44 and 45. It is hypothesized that the ester moiety acts as a highly permeable delivery vehicle and is subsequently hydrolyzed to the carboxylic acid active species by carboxylesterase enzymes. These molecules are greatly differentiated from the state-of-the-art and offer potential in the treatment of NLRP3-driven diseases, particularly where tissue penetration is required.
Collapse
Affiliation(s)
- David Harrison
- NodThera Ltd., Suite 8, The Mansion, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, United Kingdom.
| | - Nicolas Boutard
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Krzysztof Brzozka
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Marta Bugaj
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Stefan Chmielewski
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Anna Cierpich
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - John R Doedens
- NodThera Inc., 454 N 34(th) Street, Seattle, WA 98103, USA
| | | | | | - Michal Galezowski
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Piotr Kowalczyk
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Oleksandr Levenets
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Magdalena Mroczkowska
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Katarzyna Palica
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Roderick A Porter
- Rod Porter Consultancy, 89 Back Street, Ashwell, Baldock, Hertfordshire SG7 5PG, United Kingdom
| | - David Schultz
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Marta Sowinska
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Grzegorz Topolnicki
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Piotr Urbanski
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Jakub Woyciechowski
- Selvita S.A. (now Ryvu Therapeutics), Park Life Science, ul. Bobrzynskiego 14, 30-348, Kraków, Poland
| | - Alan P Watt
- NodThera Ltd., Suite 8, The Mansion, Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, United Kingdom
| |
Collapse
|
26
|
Kortylewicz ZP, Coulter DW, Han G, Baranowska-Kortylewicz J. Norepinephrine-Transporter-Targeted and DNA-Co-Targeted Theranostic Guanidines. J Med Chem 2020; 63:2051-2073. [PMID: 31268317 DOI: 10.1021/acs.jmedchem.9b00437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
High risk neuroblastoma often recurs, even with aggressive treatments. Clinical evidence suggests that proliferative activities are predictive of poor outcomes. This report describes syntheses, characterization, and biological properties of theranostic guanidines that target norepinephrine transporter and undergo intracellular processing, and subsequently their catabolites are efficiently incorporated into DNA of proliferating neuroblastoma cells. Radioactive guanidines are synthesized from 5-radioiodo-2'-deoxyuridine, a molecular radiotherapy platform with clinically proven minimal toxicities and DNA-targeting properties. The transport of radioactive guanidines into neuroblastoma cells is active as indicated by the competitive suppression of cellular uptake by meta-iodobenzylguanidine. The rate of intracellular processing and DNA uptake is influenced by the agent's catabolic stability and cell population doubling times. The radiotoxicity is directly proportional to DNA uptake and duration of exposure. Biodistribution of 5-[125I]iodo-3'-O-(ε-guanidinohexanoyl)-2'-deoxyuridine in a mouse neuroblastoma model shows significant tumor retention of radioactivity. Neuroblastoma xenografts regress in response to the clinically achievable doses of this agent.
Collapse
Affiliation(s)
- Zbigniew P Kortylewicz
- Department of Radiation Oncology, J. Bruce Henriksen Cancer Research Laboratories, University of Nebraska Medical Center, Omaha, Nebraska 68132-6850, United States
| | - Donald W Coulter
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska 68132-2168, United States
| | - Guang Han
- Department of Radiation Oncology, J. Bruce Henriksen Cancer Research Laboratories, University of Nebraska Medical Center, Omaha, Nebraska 68132-6850, United States.,Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Janina Baranowska-Kortylewicz
- Department of Radiation Oncology, J. Bruce Henriksen Cancer Research Laboratories, University of Nebraska Medical Center, Omaha, Nebraska 68132-6850, United States
| |
Collapse
|
27
|
Kundrick E, Marrero-Rosado B, Stone M, Schultz C, Walker K, Lee-Stubbs RB, de Araujo Furtado M, Lumley LA. Delayed midazolam dose effects against soman in male and female plasma carboxylesterase knockout mice. Ann N Y Acad Sci 2020; 1479:94-107. [PMID: 32027397 DOI: 10.1111/nyas.14311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/13/2020] [Accepted: 01/19/2020] [Indexed: 01/30/2023]
Abstract
Chemical warfare nerve agent exposure leads to status epilepticus that may progress to epileptogenesis and severe brain pathology when benzodiazepine treatment is delayed. We evaluated the dose-response effects of delayed midazolam (MDZ) on toxicity induced by soman (GD) in the plasma carboxylesterase knockout (Es1-/- ) mouse, which, similar to humans, lacks plasma carboxylesterase. Initially, we compared the median lethal dose (LD50 ) of GD exposure in female Es1-/- mice across estrous with male mice and observed a greater LD50 during estrus compared with proestrus or with males. Subsequently, male and female GD-exposed Es1-/- mice treated with a dose range of MDZ 40 min after seizure onset were evaluated for survivability, seizure activity, and epileptogenesis. GD-induced neuronal loss and microglial activation were evaluated 2 weeks after exposure. Similar to our previous observations in rats, delayed treatment with MDZ dose-dependently increased survival and reduced seizure severity in GD-exposed mice, but was unable to prevent epileptogenesis, neuronal loss, or gliosis. These results suggest that MDZ is beneficial against GD exposure, even when treatment is delayed, but that adjunct therapies to enhance protection need to be identified. The Es1-/- mouse GD exposure model may be useful to screen for improved medical countermeasures against nerve agent exposure.
Collapse
Affiliation(s)
- Erica Kundrick
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Brenda Marrero-Rosado
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Michael Stone
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Caroline Schultz
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Katie Walker
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Robyn B Lee-Stubbs
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | | | - Lucille A Lumley
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| |
Collapse
|
28
|
Zoubková H, Tomášková A, Nohejlová K, Černá M, Šlamberová R. Prenatal Exposure to Methamphetamine: Up-Regulation of Brain Receptor Genes. Front Neurosci 2019; 13:771. [PMID: 31417344 PMCID: PMC6686742 DOI: 10.3389/fnins.2019.00771] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/09/2019] [Indexed: 01/10/2023] Open
Abstract
Methamphetamine (METH) is a widespread illicit drug. If it is taken by pregnant women, it passes through the placenta and just as it affects the mother, it can impair the development of the offspring. The aim of our study was to identify candidates to investigate for changes in the gene expression in the specific regions of the brain associated with addiction to METH in rats. We examined the various areas of the central nervous system (striatum, hippocampus, prefrontal cortex) for signs of impairment in postnatal day 80 in experimental rats, whose mothers had been administered METH (5 mg/kg/day) during the entire gestation period. Changes in the gene expression at the mRNA level were determined by two techniques, microarray and real-time PCR. Results of two microarray trials were evaluated by LIMMA analysis. The first microarray trial detected either up-regulated or down-regulated expression of 2189 genes in the striatum; the second microarray trial detected either up-regulated or down-regulated expression of 1344 genes in the hippocampus of prenatally METH-exposed rats. We examined the expression of 10 genes using the real-time PCR technique. Differences in the gene expression were counted by the Mann–Whitney U-test. Significant changes were observed in the cocaine- and amphetamine-regulated transcript prepropeptide, tachykinin receptor 3, dopamine receptor D3 gene expression in the striatum regions, in the glucocorticoid nuclear receptor Nr3c1 gene expression in the prefrontal cortex and in the carboxylesterase 2 gene expression in the hippocampus of prenatally METH-exposed rats. The microarray technique also detected up-regulated expression of trace amine-associated receptor 7 h gene in the hippocampus of prenatally METH-exposed rats. We have identified susceptible genes; candidates for the study of an impairment related to methamphetamine addiction in the specific regions of the brain.
Collapse
Affiliation(s)
- Hana Zoubková
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Anežka Tomášková
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Kateryna Nohejlová
- Department of Physiology, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Marie Černá
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Romana Šlamberová
- Department of Physiology, Third Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
29
|
Goncharov NV, Terpilowski MA, Shmurak VI, Belinskaya DA, Avdonin PV. The Rat (Rattus norvegicus) as a Model Object for Acute Organophosphate Poisoning. 1. Biochemical Aspects. J EVOL BIOCHEM PHYS+ 2019. [DOI: 10.1134/s0022093019020042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
30
|
Marquart K, Prokopchuk O, Wilhelm D, Worek F, Thiermann H, Martignoni ME, Wille T. Human small bowel as model for poisoning with organophosphorus compounds. Toxicol In Vitro 2019; 57:76-80. [DOI: 10.1016/j.tiv.2019.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/21/2019] [Accepted: 02/09/2019] [Indexed: 11/26/2022]
|
31
|
Timperley CM, Abdollahi M, Al-Amri AS, Baulig A, Benachour D, Borrett V, Cariño FA, Geist M, Gonzalez D, Kane W, Kovarik Z, Martínez-Álvarez R, Fusaro Mourão NM, Neffe S, Raza SK, Rubaylo V, Suárez AG, Takeuchi K, Tang C, Trifirò F, van Straten FM, Vanninen PS, Vučinić S, Zaitsev V, Zafar-Uz-Zaman M, Zina MS, Holen S, Forman JE, Alwan WS, Suri V. Advice on assistance and protection by the Scientific Advisory Board of the Organisation for the Prohibition of Chemical Weapons: Part 2. On preventing and treating health effects from acute, prolonged, and repeated nerve agent exposure, and the identification of medical countermeasures able to reduce or eliminate the longer term health effects of nerve agents. Toxicology 2019; 413:13-23. [DOI: 10.1016/j.tox.2018.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/18/2018] [Accepted: 11/26/2018] [Indexed: 01/08/2023]
|
32
|
Marrero-Rosado B, de Araujo Furtado M, Schultz CR, Stone M, Kundrick E, Walker K, O'Brien S, Du F, Lumley LA. Soman-induced status epilepticus, epileptogenesis, and neuropathology in carboxylesterase knockout mice treated with midazolam. Epilepsia 2018; 59:2206-2218. [PMID: 30368799 PMCID: PMC6334636 DOI: 10.1111/epi.14582] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 12/31/2022]
Abstract
Objective Exposure to chemical warfare nerve agents (CWNAs), such as soman (GD), can induce status epilepticus (SE) that becomes refractory to benzodiazepines when treatment is delayed, leading to increased risk of epileptogenesis, severe neuropathology, and long‐term behavioral and cognitive deficits. Rodent models, widely used to evaluate novel medical countermeasures (MCMs) against CWNA exposure, normally express plasma carboxylesterase, an enzyme involved in the metabolism of certain organophosphorus compounds. To better predict the efficacy of novel MCMs against CWNA exposure in human casualties, it is crucial to use appropriate animal models that mirror the human condition. We present a comprehensive characterization of the seizurogenic, epileptogenic, and neuropathologic effects of GD exposure with delayed anticonvulsant treatment in the plasma carboxylesterase knockout (ES1−/−) mouse. Methods Electroencephalography (EEG) electrode‐implanted ES1−/− and wild‐type (C57BL/6) mice were exposed to various seizure‐inducing doses of GD, treated with atropine sulfate and the oxime HI‐6 at 1 minute after exposure, and administered midazolam at 15‐30 minutes following the onset of seizure activity. The latency of acute seizure onset and spontaneous recurrent seizures (SRS) was assessed, as were changes in EEG power spectra. At 2 weeks after GD exposure, neurodegeneration and neuroinflammation were assessed. Results GD‐exposed ES1−/− mice displayed a dose‐dependent response in seizure severity. Only ES1−/− mice exposed to the highest tested dose of GD developed SE, subchronic alterations in EEG power spectra, and SRS. Degree of neuronal cell loss and neuroinflammation were dose‐dependent; no significant neuropathology was observed in C57BL/6 mice or ES1−/− mice exposed to lower GD doses. Significance The US Food and Drug Administration (FDA) animal rule requires the use of relevant animal models for the advancement of MCMs against CWNAs. We present evidence that argues for the use of the ES1−/− mouse model to screen anticonvulsant, antiepileptic, and/or neuroprotective drugs against GD‐induced toxicity, as well as to identify mechanisms of GD‐induced epileptogenesis.
Collapse
Affiliation(s)
- Brenda Marrero-Rosado
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | | | - Caroline R Schultz
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Michael Stone
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Erica Kundrick
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Katie Walker
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Sean O'Brien
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| | - Fu Du
- FD NeuroTechnologies, Columbia, Maryland
| | - Lucille A Lumley
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, Maryland
| |
Collapse
|
33
|
Timperley CM, Forman JE, Abdollahi M, Al-Amri AS, Alonso IP, Baulig A, Borrett V, Cariño FA, Curty C, Berrutti DG, Kovarik Z, Martínez-Álvarez R, Mikulak R, Mourão NMF, Ponnadurai R, Neffe S, Raza SK, Rubaylo V, Takeuchi K, Tang C, Trifirò F, van Straten FM, Vanninen PS, Zaitsev V, Waqar F, Zina MS, Blum MM, Gregg H, Fischer E, Sun S, Yang P. Advice on chemical weapons sample stability and storage provided by the Scientific Advisory Board of the Organisation for the Prohibition of Chemical Weapons to increase investigative capabilities worldwide. Talanta 2018; 188:808-832. [DOI: 10.1016/j.talanta.2018.04.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/03/2018] [Accepted: 04/07/2018] [Indexed: 01/12/2023]
|
34
|
Marquart K, Prokopchuk O, Worek F, Thiermann H, Martignoni ME, Wille T. Human small bowel as a useful tool to investigate smooth muscle effects of potential therapeutics in organophosphate poisoning. Toxicol Lett 2018; 293:235-240. [PMID: 29154801 DOI: 10.1016/j.toxlet.2017.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/07/2017] [Accepted: 11/12/2017] [Indexed: 11/29/2022]
Abstract
Isolated organs proofed to be a robust tool to study effects of (potential) therapeutics in organophosphate poisoning. Small bowel samples have been successfully used to reveal smooth muscle relaxing effects. In the present study, the effects of obidoxime, TMB-4, HI-6 and MB 327 were investigated on human small bowel tissue and compared with rat data. Hereby, the substances were tested in at least seven different concentrations in the jejunum or ileum both pre-contracted with carbamoylcholine. Additionally, the cholinesterase activity of native tissue was determined. Human small intestine specimens showed classical dose response-curves, similar to rat tissue, with MB 327 exerting the most potent smooth muscle relaxant effect in both species (human EC50=0.7×10-5M and rat EC50=0.7×10-5M). The AChE activity for human and rat samples did not differ significantly (rat jejunum=1351±166 mU/mg wet weight; rat ileum=1078±123 mU/mg wet weight; human jejunum=1030±258 mU/mg wet weight; human ileum=1293±243 mU/mg wet weight). Summarizing, our isolated small bowel setup seems to be a solid tool to investigate the effects of (potential) therapeutics on pre-contracted smooth muscle, with data being transferable between rat and humans.
Collapse
Affiliation(s)
- Katharina Marquart
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80397 Munich, Germany
| | - Olga Prokopchuk
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675 Munich, Germany
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80397 Munich, Germany
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80397 Munich, Germany
| | - Marc E Martignoni
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675 Munich, Germany
| | - Timo Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstr. 11, 80397 Munich, Germany.
| |
Collapse
|
35
|
Catalytic bioscavengers as countermeasures against organophosphate nerve agents. Chem Biol Interact 2018; 292:50-64. [DOI: 10.1016/j.cbi.2018.07.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 12/30/2022]
|
36
|
Dunn EN, Ferrara-Bowens TM, Chachich ME, Honnold CL, Rothwell CC, Hoard-Fruchey HM, Lesyna CA, Johnson EA, Cerasoli DM, McDonough JH, Cadieux CL. Evaluating mice lacking serum carboxylesterase as a behavioral model for nerve agent intoxication. Toxicol Mech Methods 2018; 28:563-572. [PMID: 29768075 DOI: 10.1080/15376516.2018.1476637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mice and other rodents are typically utilized for chemical warfare nerve agent research. Rodents have large amounts of carboxylesterase in their blood, while humans do not. Carboxylesterase nonspecifically binds to and detoxifies nerve agent. The presence of this natural bioscavenger makes mice and other rodents poor models for studies identifying therapeutics to treat humans exposed to nerve agents. To obviate this problem, a serum carboxylesterase knockout (Es1 KO) mouse was created. In this study, Es1 KO and wild type (WT) mice were assessed for differences in gene expression, nerve agent (soman; GD) median lethal dose (MLD) values, and behavior prior to and following nerve agent exposure. No expression differences were detected between Es1 KO and WT mice in more than 34 000 mouse genes tested. There was a significant difference between Es1 KO and WT mice in MLD values, as the MLD for GD-exposed WT mice was significantly higher than the MLD for GD-exposed Es1 KO mice. Behavioral assessments of Es1 KO and WT mice included an open field test, a zero maze, a Barnes maze, and a sucrose preference test (SPT). While sex differences were observed in various measures of these tests, overall, Es1 KO mice behaved similarly to WT mice. The two genotypes also showed virtually identical neuropathological changes following GD exposure. Es1 KO mice appear to have an enhanced susceptibility to GD toxicity while retaining all other behavioral and physiological responses to this nerve agent, making the Es1 KO mouse a more human-like model for nerve agent research.
Collapse
Affiliation(s)
- Emily N Dunn
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Teresa M Ferrara-Bowens
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Mark E Chachich
- b Department of Psychology , Towson University , Towson , MD , USA
| | - Cary L Honnold
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Cristin C Rothwell
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Heidi M Hoard-Fruchey
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Catherine A Lesyna
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Erik A Johnson
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - Douglas M Cerasoli
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - John H McDonough
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| | - C Linn Cadieux
- a United States Army Medical Research Institute of Chemical Defense , Aberdeen Proving Ground , MD , USA
| |
Collapse
|
37
|
The role of genetic background in susceptibility to chemical warfare nerve agents across rodent and non-human primate models. Toxicology 2017; 393:51-61. [PMID: 29113833 DOI: 10.1016/j.tox.2017.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/13/2017] [Accepted: 11/02/2017] [Indexed: 11/22/2022]
Abstract
Genetics likely play a role in various responses to nerve agent exposure, as genetic background plays an important role in behavioral, neurological, and physiological responses to environmental stimuli. Mouse strains or selected lines can be used to identify susceptibility based on background genetic features to nerve agent exposure. Additional genetic techniques can then be used to identify mechanisms underlying resistance and sensitivity, with the ultimate goal of developing more effective and targeted therapies. Here, we discuss the available literature on strain and selected line differences in cholinesterase activity levels and response to nerve agent-induced toxicity and seizures. We also discuss the available cholinesterase and toxicity literature across different non-human primate species. The available data suggest that robust genetic differences exist in cholinesterase activity, nerve agent-induced toxicity, and chemical-induced seizures. Available cholinesterase data suggest that acetylcholinesterase activity differs across strains, but are limited by the paucity of carboxylesterase data in strains and selected lines. Toxicity and seizures, two outcomes of nerve agent exposure, have not been fully evaluated for genetic differences, and thus further studies are required to understand baseline strain and selected line differences.
Collapse
|
38
|
Makhaeva GF, Rudakova EV, Serebryakova OG, Aksinenko AY, Lushchekina SV, Bachurin SO, Richardson RJ. Esterase profiles of organophosphorus compounds in vitro predict their behavior in vivo. Chem Biol Interact 2016; 259:332-342. [DOI: 10.1016/j.cbi.2016.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 10/21/2022]
|
39
|
Makhaeva GF, Rudakova EV, Serebryakova OG, Aksinenko AY, Richardson RJ, Bachurin SO. Esterase profiles of hexafluoropropan-2-ol-based dialkyl phosphates as a major determinant of their effects in mouse brain in vivo. Russ Chem Bull 2016. [DOI: 10.1007/s11172-015-1139-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
40
|
Naturally Occurring Genetic Variants of Human Acetylcholinesterase and Butyrylcholinesterase and Their Potential Impact on the Risk of Toxicity from Cholinesterase Inhibitors. Chem Res Toxicol 2016; 29:1381-92. [PMID: 27551784 PMCID: PMC5030680 DOI: 10.1021/acs.chemrestox.6b00228] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
![]()
Acetylcholinesterase
(AChE) is the physiologically important target
for organophosphorus toxicants (OP) including nerve agents and pesticides.
Butyrylcholinesterase (BChE) in blood serves as a bioscavenger that
protects AChE in nerve synapses from inhibition by OP. Mass spectrometry
methods can detect exposure to OP by measuring adducts on the active
site serine of plasma BChE. Genetic variants of human AChE and BChE
do exist, but loss of function mutations have been identified only
in the BCHE gene. The most common AChE variant, His353Asn (H322N),
also known as the Yt blood group antigen, has normal AChE activity.
The most common BChE variant, Ala567Thr (A539T) or the K-variant in
honor of Werner Kalow, has 33% reduced plasma BChE activity. The genetic
variant most frequently associated with prolonged response to muscle
relaxants, Asp98Gly (D70G) or atypical BChE, has reduced activity
and reduced enzyme concentration. Early studies in young, healthy
males, performed at a time when it was legal to test nerve agents
in humans, showed that individuals responded differently to the same
low dose of sarin with toxic symptoms ranging in severity from minimal
to moderate. Additionally, animal studies indicated that BChE protects
from toxicants that have a higher reactivity with AChE than with BChE
(e.g., nerve agents) but not from toxicants that have a higher reactivity
with BChE than with AChE (e.g., OP pesticides). As a corollary, we
hypothesize that individuals with genetic variants of BChE may be
at increased risk of toxicity from nerve agents but not from OP pesticides.
Collapse
|
41
|
Dorywalska M, Dushin R, Moine L, Farias SE, Zhou D, Navaratnam T, Lui V, Hasa-Moreno A, Casas MG, Tran TT, Delaria K, Liu SH, Foletti D, O'Donnell CJ, Pons J, Shelton DL, Rajpal A, Strop P. Molecular Basis of Valine-Citrulline-PABC Linker Instability in Site-Specific ADCs and Its Mitigation by Linker Design. Mol Cancer Ther 2016; 15:958-70. [PMID: 26944918 DOI: 10.1158/1535-7163.mct-15-1004] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 02/12/2016] [Indexed: 11/16/2022]
Abstract
The degree of stability of antibody-drug linkers in systemic circulation, and the rate of their intracellular processing within target cancer cells are among the key factors determining the efficacy of antibody-drug conjugates (ADC) in vivo Previous studies demonstrated the susceptibility of cleavable linkers, as well as auristatin-based payloads, to enzymatic cleavage in rodent plasma. Here, we identify Carboxylesterase 1C as the enzyme responsible for the extracellular hydrolysis of valine-citrulline-p-aminocarbamate (VC-PABC)-based linkers in mouse plasma. We further show that the activity of Carboxylesterase 1C towards VC-PABC-based linkers, and consequently the stability of ADCs in mouse plasma, can be effectively modulated by small chemical modifications to the linker. While the introduced modifications can protect the VC-PABC-based linkers from extracellular cleavage, they do not significantly alter the intracellular linker processing by the lysosomal protease Cathepsin B. The distinct substrate preference of the serum Carboxylesterase 1C offers the opportunity to modulate the extracellular stability of cleavable ADCs without diminishing the intracellular payload release required for ADC efficacy. Mol Cancer Ther; 15(5); 958-70. ©2016 AACR.
Collapse
Affiliation(s)
| | - Russell Dushin
- Worldwide Medicinal Chemistry, Pfizer Inc., Groton, Connecticut
| | - Ludivine Moine
- Worldwide Medicinal Chemistry, Pfizer Inc., Groton, Connecticut
| | | | - Dahui Zhou
- Worldwide Medicinal Chemistry, Pfizer Inc., Groton, Connecticut
| | | | - Victor Lui
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | | | | | - Thomas-Toan Tran
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | - Kathy Delaria
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | - Shu-Hui Liu
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | - Davide Foletti
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | | | - Jaume Pons
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | - David L Shelton
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | - Arvind Rajpal
- Rinat Laboratories, Pfizer Inc., South San Francisco, California
| | - Pavel Strop
- Rinat Laboratories, Pfizer Inc., South San Francisco, California.
| |
Collapse
|
42
|
Purification of a chymotrypsin-like enzyme present on adult Schistosoma mansoni worms from infected mice and its characterization as a host carboxylesterase. Parasitology 2016; 143:646-57. [DOI: 10.1017/s0031182016000184] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SUMMARYA serine protease-like enzyme found in detergent extracts of Schistosoma mansoni adult worms perfused from infected mice has been purified from mouse blood and further characterized. The enzyme is approximately 85 kDa and hydrolyses N-acetyl-DL-phenylalanine β-naphthyl–ester, a chromogenic substrate for chymotrypsin-like enzymes. The enzyme from S. mansoni worms appears to be antigenically and enzymatically similar to a molecule that is present in normal mouse blood and so is seemingly host-derived. The enzyme was partially purified by depleting normal mouse serum of albumin using sodium chloride and cold ethanol, followed by repeated rounds of purification by one-dimensional sodium dodecyl sulphate polyacrylamide gel electrophoresis. The purified material was subjected to tandem mass spectrometry and its derived peptides found to belong to mouse carboxylesterase 1C. Its ability to hydrolyse α- or β-naphthyl acetates, which are general esterase substrates, has been confirmed. A similar carboxylesterase was purified and characterized from rat blood. Additional evidence to support identification of the enzyme as a carboxylesterase has been provided. Possible roles of the enzyme in the mouse host–parasite relationship could be to ease the passage of worms through the host's blood vessels and/or in immune evasion.
Collapse
|
43
|
Peng H, Brimijoin S, Hrabovska A, Krejci E, Blake TA, Johnson RC, Masson P, Lockridge O. Monoclonal antibodies to human butyrylcholinesterase reactive with butyrylcholinesterase in animal plasma. Chem Biol Interact 2015; 243:82-90. [PMID: 26585590 DOI: 10.1016/j.cbi.2015.11.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/01/2015] [Accepted: 11/09/2015] [Indexed: 11/18/2022]
Abstract
Five mouse anti-human butyrylcholinesterase (BChE) monoclonal antibodies bind tightly to native human BChE with nanomolar dissociation constants. Pairing analysis in the Octet system identified the monoclonal antibodies that bind to overlapping and independent epitopes on human BChE. The nucleotide and amino acid sequences of 4 monoclonal antibodies are deposited in GenBank. Our goal was to determine which of the 5 monoclonal antibodies recognize BChE in the plasma of animals. Binding of monoclonal antibodies 11D8, B2 18-5, B2 12-1, mAb2 and 3E8 to BChE in animal plasma was measured using antibody immobilized on Pansorbin cells and on Dynabeads Protein G. A third method visualized binding by the shift of BChE activity bands on nondenaturing gels stained for BChE activity. Gels were counterstained for carboxylesterase activity. The three methods agreed that B2 18-5 and mAb2 have broad species specificity, but the other monoclonal antibodies interacted only with human BChE, the exception being 3E8, which also bound chicken BChE. B2 18-5 and mAb2 recognized BChE in human, rhesus monkey, horse, cat, and tiger plasma. A weak response was found with rabbit BChE. Monoclonal mAb2, but not B2 18-5, bound pig and bovine BChE. Gels stained for carboxylesterase activity confirmed that plasma from humans, monkey, pig, chicken, and cow does not contain carboxylesterase, but plasma from horse, cat, tiger, rabbit, guinea pig, mouse, and rat has carboxylesterase. Rabbit plasma carboxylesterase hydrolyzes butyrylthiocholine. In conclusion monoclonal antibodies B2 18-5 and mAb2 can be used to immuno extract BChE from the plasma of humans, monkey and other animals.
Collapse
Affiliation(s)
- Hong Peng
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Stephen Brimijoin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Anna Hrabovska
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia.
| | - Eric Krejci
- Université Paris Descartes CNRS SSA COGNAC G UMR 8257 45 rue des Saints Pères, 75006, Paris, France.
| | - Thomas A Blake
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, 30341, USA.
| | - Rudolph C Johnson
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, 30341, USA.
| | - Patrick Masson
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Oksana Lockridge
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
44
|
Katz FS, Pecic S, Tran TH, Trakht I, Schneider L, Zhu Z, Ton-That L, Luzac M, Zlatanic V, Damera S, Macdonald J, Landry DW, Tong L, Stojanovic MN. Discovery of New Classes of Compounds that Reactivate Acetylcholinesterase Inhibited by Organophosphates. Chembiochem 2015; 16:2205-2215. [PMID: 26350723 DOI: 10.1002/cbic.201500348] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Indexed: 11/11/2022]
Abstract
Acetylcholinesterase (AChE) that has been covalently inhibited by organophosphate compounds (OPCs), such as nerve agents and pesticides, has traditionally been reactivated by using nucleophilic oximes. There is, however, a clearly recognized need for new classes of compounds with the ability to reactivate inhibited AChE with improved in vivo efficacy. Here we describe our discovery of new functional groups--Mannich phenols and general bases--that are capable of reactivating OPC--inhibited AChE more efficiently than standard oximes and we describe the cooperative mechanism by which these functionalities are delivered to the active site. These discoveries, supported by preliminary in vivo results and crystallographic data, significantly broaden the available approaches for reactivation of AChE.
Collapse
Affiliation(s)
- Francine S Katz
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA)
| | - Stevan Pecic
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA)
| | - Timothy H Tran
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Avenue, New York, NY 10027 (USA)
| | - Ilya Trakht
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA)
| | - Laura Schneider
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA)
| | - Zhengxiang Zhu
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA)
| | - Long Ton-That
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA)
| | - Michal Luzac
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA)
| | - Viktor Zlatanic
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA)
| | - Shivani Damera
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA)
| | - Joanne Macdonald
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA).,Genecology Research Centre, Inflammation and Healing Research Cluster, School of Science and Engineering, University of the Sunshine Coast, 90 Sippy Downs Drive, Sippy Downs, QLD 4556 (Australia)
| | - Donald W Landry
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA)
| | - Liang Tong
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Avenue, New York, NY 10027 (USA)
| | - Milan N Stojanovic
- Department of Medicine/Division of Experimental Therapeutics, Columbia University Medical Center, 630 W. 168th Street, New York, NY 10032 (USA).,Departments of Biomedical Engineering and Systems Biology, Columbia University, 630 W. 168th street, New York, NY 10032 (USA)
| |
Collapse
|
45
|
Abstract
Ongoing mouse studies of a proposed therapy for cocaine abuse based on viral gene transfer of butyrylcholinesterase (BChE) mutated for accelerated cocaine hydrolysis have yielded surprising effects on aggression. Further investigation has linked these effects to a reduction in circulating ghrelin, driven by BChE at levels ∼ 100-fold above normal. Tests with human BChE showed ready ghrelin hydrolysis at physiologic concentrations, and multiple low-mass molecular dynamics simulations revealed that ghrelin's first five residues fit sterically and electrostatically into BChE's active site. Consistent with in vitro results, male BALB/c mice with high plasma BChE after gene transfer exhibited sharply reduced plasma ghrelin. Unexpectedly, such animals fought less, both spontaneously and in a resident/intruder provocation model. One mutant BChE was found to be deficient in ghrelin hydrolysis. BALB/c mice transduced with this variant retained normal plasma ghrelin levels and did not differ from untreated controls in the aggression model. In contrast, C57BL/6 mice with BChE gene deletion exhibited increased ghrelin and fought more readily than wild-type animals. Collectively, these findings indicate that BChE-catalyzed ghrelin hydrolysis influences mouse aggression and social stress, with potential implications for humans.
Collapse
|
46
|
Oda S, Fukami T, Yokoi T, Nakajima M. A comprehensive review of UDP-glucuronosyltransferase and esterases for drug development. Drug Metab Pharmacokinet 2015; 30:30-51. [DOI: 10.1016/j.dmpk.2014.12.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/24/2014] [Accepted: 12/02/2014] [Indexed: 01/24/2023]
|
47
|
van der Lee MMC, Groothuis PG, Ubink R, van der Vleuten MAJ, van Achterberg TA, Loosveld EM, Damming D, Jacobs DCH, Rouwette M, Egging DF, van den Dobbelsteen D, Beusker PH, Goedings P, Verheijden GFM, Lemmens JM, Timmers M, Dokter WHA. The Preclinical Profile of the Duocarmycin-Based HER2-Targeting ADC SYD985 Predicts for Clinical Benefit in Low HER2-Expressing Breast Cancers. Mol Cancer Ther 2015; 14:692-703. [PMID: 25589493 DOI: 10.1158/1535-7163.mct-14-0881-t] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/22/2014] [Indexed: 11/16/2022]
Abstract
SYD985 is a HER2-targeting antibody-drug conjugate (ADC) based on trastuzumab and vc-seco-DUBA, a cleavable linker-duocarmycin payload. To evaluate the therapeutic potential of this new ADC, mechanistic in vitro studies and in vivo patient-derived xenograft (PDX) studies were conducted to compare SYD985 head-to-head with T-DM1 (Kadcyla), another trastuzumab-based ADC. SYD985 and T-DM1 had similar binding affinities to HER2 and showed similar internalization. In vitro cytotoxicity assays showed similar potencies and efficacies in HER2 3+ cell lines, but in cell lines with low HER2 expression, SYD985 was 3- to 50-fold more potent than T-DM1. In contrast with T-DM1, SYD985 efficiently induced bystander killing in vitro in HER2-negative (HER2 0) cells mixed with HER2 3+, 2+, or 1+ cell lines. At pH conditions relevant for tumors, cathepsin-B cleavage studies showed efficient release of the active toxin by SYD985 but not by T-DM1. These in vitro data suggest that SYD985 might be a more potent ADC in HER2-expressing tumors in vivo, especially in low HER2-expressing and/or in heterogeneous tumors. In line with this, in vivo antitumor studies in breast cancer PDX models showed that SYD985 is very active in HER2 3+, 2+, and 1+ models, whereas T-DM1 only showed significant antitumor activity in HER2 3+ breast cancer PDX models. These properties of SYD985 may enable expansion of the target population to patients who have low HER2-expressing breast cancer, a patient population with still unmet high medical need.
Collapse
Affiliation(s)
| | - Patrick G Groothuis
- Department of Preclinical, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | - Ruud Ubink
- Department of Preclinical, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | | | | | - Eline M Loosveld
- Department of Preclinical, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | - Désirée Damming
- Department of Preclinical, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | - Daniëlle C H Jacobs
- Department of Preclinical, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | - Myrthe Rouwette
- Department of Preclinical, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | - David F Egging
- Department of Preclinical, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | | | - Patrick H Beusker
- Department of Medicinal and Protein Chemistry, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | - Peter Goedings
- Department of Medical R&D, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | - Gijs F M Verheijden
- Department of New Molecular Entities, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | - Jacques M Lemmens
- Department of New Molecular Entities, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | - Marco Timmers
- Department of New Molecular Entities, Synthon Biopharmaceuticals, Nijmegen, the Netherlands
| | - Wim H A Dokter
- Department of Preclinical, Synthon Biopharmaceuticals, Nijmegen, the Netherlands.
| |
Collapse
|
48
|
Wong CC, Cheng KW, Papayannis I, Mattheolabakis G, Huang L, Xie G, Ouyang N, Rigas B. Phospho-NSAIDs have enhanced efficacy in mice lacking plasma carboxylesterase: implications for their clinical pharmacology. Pharm Res 2014; 32:1663-75. [PMID: 25392229 DOI: 10.1007/s11095-014-1565-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 10/28/2014] [Indexed: 12/31/2022]
Abstract
PURPOSE The purpose of the study was to evaluate the metabolism, pharmacokinetics and efficacy of phospho-NSAIDs in Ces1c-knockout mice. METHODS Hydrolysis of phospho-NSAIDs by Ces1c was investigated using Ces1c-overexpressing cells. The rate of phospho-NSAID hydrolysis was compared between wild-type, Ces1c+/- and Ces1c-/- mouse plasma in vitro, and the effect of plasma Ces1c on the cytotoxicity of phospho-NSAIDs was evaluated. Pharmacokinetics of phospho-sulindac was examined in wild-type and Ces1c-/- mice. The impact of Ces1c on the efficacy of phospho-sulindac was investigated using lung and pancreatic cancer models in vivo. RESULTS Phospho-NSAIDs were extensively hydrolyzed in Ces1c-overexpressing cells. Phospho-NSAID hydrolysis in wild-type mouse plasma was 6-530-fold higher than that in the plasma of Ces1c-/- mice. Ces1c-expressing wild-type mouse serum attenuated the in vitro cytotoxicity of phospho-NSAIDs towards cancer cells. Pharmacokinetic studies of phospho-sulindac using wild-type and Ces1c-/- mice demonstrated 2-fold less inactivation of phospho-sulindac in the latter. Phospho-sulindac was 2-fold more efficacious in inhibiting the growth of lung and pancreatic carcinoma in Ces1c -/- mice, as compared to wild-type mice. CONCLUSIONS Our results indicate that intact phospho-NSAIDs are the pharmacologically active entities and phospho-NSAIDs are expected to be more efficacious in humans than in rodents due to their differential expression of carboxylesterases.
Collapse
Affiliation(s)
- Chi C Wong
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, HSC, T-17 Room 080, Stony Brook, NY, 11794-8173, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Dokter W, Ubink R, van der Lee M, van der Vleuten M, van Achterberg T, Jacobs D, Loosveld E, van den Dobbelsteen D, Egging D, Mattaar E, Groothuis P, Beusker P, Coumans R, Elgersma R, Menge W, Joosten J, Spijker H, Huijbregts T, de Groot V, Eppink M, de Roo G, Verheijden G, Timmers M. Preclinical Profile of the HER2-Targeting ADC SYD983/SYD985: Introduction of a New Duocarmycin-Based Linker-Drug Platform. Mol Cancer Ther 2014; 13:2618-29. [DOI: 10.1158/1535-7163.mct-14-0040-t] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
50
|
Rudakova EV, Makhaeva GF, Galenko TG, Aksinenko AY, Sokolov VB, Martynov IV. A new selective inhibitor of mouse blood plasma carboxylesterase. DOKL BIOCHEM BIOPHYS 2013; 449:87-9. [DOI: 10.1134/s1607672913020099] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Indexed: 11/23/2022]
|