Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where David L. Linemeyer is active.

Publication


Featured researches published by David L. Linemeyer.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Targeting thyroid hormone receptor-beta agonists to the liver reduces cholesterol and triglycerides and improves the therapeutic index.

Mark D. Erion; Ee Cable; Bruce R. Ito; Hongjian Jiang; James M. Fujitaki; Patricia D. Finn; Bao-Hong Zhang; Jinzhao Hou; Serge H. Boyer; Paul D. van Poelje; David L. Linemeyer

Despite efforts spanning four decades, the therapeutic potential of thyroid hormone receptor (TR) agonists as lipid-lowering and anti-obesity agents remains largely unexplored in humans because of dose-limiting cardiac effects and effects on the thyroid hormone axis (THA), muscle metabolism, and bone turnover. TR agonists selective for the TRβ isoform exhibit modest cardiac sparing in rodents and primates but are unable to lower lipids without inducing TRβ-mediated suppression of the THA. Herein, we describe a cytochrome P450-activated prodrug of a phosphonate-containing TR agonist that exhibits increased TR activation in the liver relative to extrahepatic tissues and an improved therapeutic index. Pharmacokinetic studies in rats demonstrated that the prodrug (2R,4S)-4-(3-chlorophenyl)-2-[(3,5-dimethyl-4-(4′-hydroxy-3′-isopropylbenzyl)phenoxy)methyl]-2-oxido-[1,3,2]-dioxaphosphonane (MB07811) undergoes first-pass hepatic extraction and that cleavage of the prodrug generates the negatively charged TR agonist (3,5-dimethyl-4-(4′-hydroxy-3′-isopropylbenzyl)phenoxy)methylphosphonic acid (MB07344), which distributes poorly into most tissues and is rapidly eliminated in the bile. Enhanced liver targeting was further demonstrated by comparing the effects of MB07811 with 3,5,3′-triiodo-l-thyronine (T3) and a non-liver-targeted TR agonist, 3,5-dichloro-4-(4-hydroxy-3-isopropylphenoxy)phenylacetic acid (KB-141) on the expression of TR agonist-responsive genes in the liver and six extrahepatic tissues. The pharmacologic effects of liver targeting were evident in the normal rat, where MB07811 exhibited increased cardiac sparing, and in the diet-induced obese mouse, where, unlike KB-141, MB07811 reduced cholesterol and both serum and hepatic triglycerides at doses devoid of effects on body weight, glycemia, and the THA. These results indicate that targeting TR agonists to the liver has the potential to lower both cholesterol and triglyceride levels with an acceptable safety profile.


Journal of Pharmacology and Experimental Therapeutics | 2004

Liver-Targeted Drug Delivery Using HepDirect Prodrugs

Mark D. Erion; Paul D. van Poelje; Deidre A. MacKenna; Timothy J. Colby; Annika C. Montag; James M. Fujitaki; David L. Linemeyer; David A. Bullough

Targeting drugs to specific organs, tissues, or cells is an attractive strategy for enhancing drug efficacy and reducing side effects. Drug carriers such as antibodies, natural and manmade polymers, and labeled liposomes are capable of targeting drugs to blood vessels of individual tissues but often fail to deliver drugs to extravascular sites. An alternative strategy is to use low molecular weight prodrugs that distribute throughout the body but cleave intracellularly to the active drug by an organ-specific enzyme. Here we show that a series of phosphate and phosphonate prodrugs, called HepDirect prodrugs, results in liver-targeted drug delivery following a cytochrome P450-catalyzed oxidative cleavage reaction inside hepatocytes. Liver targeting was demonstrated in rodents for MB06866 [(2R,4S)-9-[2-[4-(3-chlorophenyl)-2-oxo-1,3,2-dioxaphosphorinan-2-yl]methoxyethyl]adenine (remofovir)], a Hep-Direct prodrug of the nucleotide analog adefovir (PMEA), and MB07133 [(2R,4S)-4-amino-1-[5-O-[2-oxo-4-(4-pyridyl)-1,3,2-dioxaphosphorinan-2-yl]-β-d-arabinofuranosyl]-2(1H)-pyrimidinone], a HepDirect prodrug of cytarabine (araC) 5′-monophosphate. Liver targeting led to higher levels of the biologically active form of PMEA and araC in the liver and to lower levels in the most toxicologically sensitive organs. Liver targeting also confined production of the prodrug byproduct, an aryl vinyl ketone, to hepatocytes. Glutathione within the hepatocytes rapidly reacted with the byproduct to form a glutathione conjugate. No byproduct-related toxicity was observed in hepatocytes or animals treated with HepDirect prodrugs. A 5-day safety study in mice demonstrated the toxicological benefits of liver targeting. These findings suggest that HepDirect prodrugs represent a potential strategy for targeting drugs to the liver and achieving more effective therapies against chronic liver diseases such as hepatitis B, hepatitis C, and hepatocellular carcinoma.


Hepatology | 2009

Reduction of hepatic steatosis in rats and mice after treatment with a liver‐targeted thyroid hormone receptor agonist

Edward E. Cable; Patricia D. Finn; Jeffrey W. Stebbins; Jinzhao Hou; Bruce R. Ito; Paul D. van Poelje; David L. Linemeyer; Mark D. Erion

Non‐alcoholic fatty liver disease (NAFLD) is one of the most common forms of chronic liver disease, with a prevalence ranging from 10% to 30%. The use of thyroid hormone receptor (TR) agonists for the treatment of NAFLD has not been considered viable because thyroid hormones increase free fatty acid (FFA) flux from the periphery to the liver, induce hepatic lipogenesis, and therefore could potentially cause steatosis. MB07811 is an orally active HepDirect prodrug of MB07344, a liver‐targeted TR‐β agonist. The purpose of these studies was to assess the effects of MB07811 on whole body and liver lipid metabolism of normal rodents and rodent models of hepatic steatosis. In the current studies, MB07811 markedly reduced hepatic steatosis as well as reduced plasma FFA and triglycerides. In contrast to MB07811, T3 induced adipocyte lipolysis in vitro and in vivo and had a diminished ability to decrease hepatic steatosis. This suggests the influx of FFA from the periphery to the liver may partially counteract the antisteatotic activity of T3. Clearance of liver lipids by MB07811 results from accelerated hepatic fatty acid oxidation, a known consequence of hepatic TR activation, as reflected by increased hepatic mitochondrial respiration rates, changes in hepatic gene expression, and increased plasma acyl‐carnitine levels. Transaminase levels remained unchanged, or were reduced, and no evidence for liver fibrosis or other histological liver damage was observed after treatment with MB07811 for up to 10 weeks. Additionally, MB07811, unlike T3, did not increase heart weight or decrease pituitary thyroid‐stimulating hormone beta (TSHβ) expression. Conclusion: MB07811 represents a novel class of liver‐targeted TR agonists with beneficial low‐density lipoprotein cholesterol‐lowering properties that may provide additional therapeutic benefit to hyperlipidemic patients with concomitant NAFLD. (HEPATOLOGY 2009.)


ACS Medicinal Chemistry Letters | 2010

A Potent and Selective AMPK Activator That Inhibits de Novo Lipogenesis

Jorge E. Gomez-Galeno; Qun Dang; Thanh Huu Nguyen; Serge H. Boyer; Matthew P. Grote; Zhili Sun; Mingwei Chen; William Craigo; Paul D. van Poelje; Deidre A. MacKenna; Edward E. Cable; Paul A. Rolzin; Patricia D. Finn; Bert Chi; David L. Linemeyer; Scott J. Hecker; Mark D. Erion

AMP-activated protein kinase (AMPK) is a heterotrimeric kinase that regulates cellular energy metabolism by affecting energy-consuming pathways such as de novo lipid biosynthesis and glucose production as well as energy-producing pathways such as lipid oxidation and glucose uptake. Accordingly, compounds that activate AMPK represent potential drug candidates for the treatment of hyperlipidemia and type 2 diabetes. Screening of a proprietary library of AMP mimetics identified the phosphonic acid 2 that bears little structural resemblance to AMP but is capable of activating AMPK with high potency (EC50 = 6 nM vs AMP EC50 = 6 μM) and specificity. Phosphonate prodrugs of 2 inhibited de novo lipogenesis in cellular and animal models of hyperlipidemia.


British Journal of Pharmacology | 2009

Thyroid hormone β receptor activation has additive cholesterol lowering activity in combination with atorvastatin in rabbits, dogs and monkeys

Bruce R. Ito; B-H Zhang; Ee Cable; X Song; James M. Fujitaki; Deidre A. MacKenna; Ce Wilker; B Chi; Pd van Poelje; David L. Linemeyer; Erion

Background and purpose:u2002 Thyroid hormone receptor (TR) agonists are in clinical trials for the treatment of hypercholesterolaemia. As statins are the standard of clinical care, any new therapies must have adjunctive activity, when given in combination with statins. As already known for the statins, the cholesterol lowering effect of TR activation involves increased expression of the low‐density lipoprotein receptor. Using animal models, we tested whether TR activation would have additive cholesterol lowering activity in the presence of effective doses of a statin.


Drug Metabolism and Disposition | 2008

Preclinical pharmacokinetics of a HepDirect prodrug of a novel phosphonate-containing thyroid hormone receptor agonist.

James M. Fujitaki; Edward E. Cable; Bruce R. Ito; Bao-Hong Zhang; Jinzhao Hou; Chun Yang; David A. Bullough; James L. Ferrero; Paul D. van Poelje; David L. Linemeyer; Mark D. Erion

The prodrug [(2R,4S)-4-(3-chlorophenyl)-2-[(3,5-dimethyl-4-(4′-hydroxy-3′-isopropylbenzyl)phenoxy)methyl]-2-oxido-[1,3,2]-dioxaphosphonane (MB07811)] of a novel phosphonate-containing thyroid hormone receptor agonist [3,5-dimethyl-4-(4′-hydroxy-3′-isopropylbenzyl)phenoxylmethylphosphonic acid (MB07344)] is the first application of the HepDirect liver-targeting approach to a non-nucleotide agent. The disposition of MB07811 was characterized in rat, dog, and monkey to assess its liver specificity, which is essential in limiting the extrahepatic side effects associated with this class of lipid-lowering agents. MB07811 was converted to MB07344 in liver microsomes from all species tested (CLint 1.23-145.4 μl/min/mg). The plasma clearance and volume of distribution of MB07811 matched or exceeded 1 l/h/kg and 3 l/kg, respectively. Although absorption of prodrug was good, its absolute oral bioavailability as measured systemically was low (3-10%), an indication of an extensive hepatic first-pass effect. This effect was confirmed by comparison of systemic exposure levels of MB07811 after portal and jugular vein administration to rats, which demonstrated a hepatic extraction ratio of >0.6 with liver CYP3A-mediated conversion to MB07344 being a major component. The main route of elimination of MB07811 and MB07344 was biliary, with no evidence for enterohepatic recirculation of MB07344. Similar metabolic profiles of MB07811 were obtained in liver microsomes across the species tested. Tissue distribution and whole body autoradiography confirmed that the liver is the major target organ of MB07811 and that conversion to MB07344 was high in the liver relative to that in other tissues. Hepatic first-pass extraction and metabolism of MB07811, coupled with possible selective distribution of MB07811-derived MB07344, led to a high degree of liver targeting of MB07344.


Antimicrobial Agents and Chemotherapy | 2011

Antiviral Efficacy upon Administration of a HepDirect Prodrug of 2′-C-Methylcytidine to Hepatitis C Virus-Infected Chimpanzees

Steven S. Carroll; Kenneth A. Koeplinger; Marissa Vavrek; Nanyan Rena Zhang; Laurence Handt; Malcolm MacCoss; David B. Olsen; K. Raja Reddy; Zhili Sun; Paul D. van Poelje; James M. Fujitaki; Serge H. Boyer; David L. Linemeyer; Scott J. Hecker; Mark D. Erion

ABSTRACT Hepatitis C virus (HCV) infects an estimated 170 million individuals worldwide, and the current standard of care, a combination of pegylated interferon alpha and ribavirin, is efficacious in achieving sustained viral response in ∼50% of treated patients. Novel therapies under investigation include the use of nucleoside analog inhibitors of the viral RNA-dependent RNA polymerase. NM283, a 3′-valyl ester prodrug of 2′-C-methylcytidine, has demonstrated antiviral efficacy in HCV-infected patients (N. Afdhal et al., J. Hepatol. 46[Suppl. 1]:S5, 2007; N. Afdhal et al., J. Hepatol. 44[Suppl. 2]:S19, 2006). One approach to increase the antiviral efficacy of 2′-C-methylcytidine is to increase the concentration of the active inhibitory species, the 5′-triphosphate, in infected hepatocytes. HepDirect prodrug technology can increase intracellular concentrations of a nucleoside triphosphate in hepatocytes by introducing the nucleoside monophosphate into the cell, bypassing the initial kinase step that is often rate limiting. Screening for 2′-C-methylcytidine triphosphate levels in rat liver after oral dosing identified 1-[3,5-difluorophenyl]-1,3-propandiol as an efficient prodrug modification. To determine antiviral efficacy in vivo, the prodrug was administered separately via oral and intravenous dosing to two HCV-infected chimpanzees. Circulating viral loads declined by ∼1.4 log10 IU/ml and by >3.6 log10 IU/ml after oral and intravenous dosing, respectively. The viral loads rebounded after the end of dosing to predose levels. The results indicate that a robust antiviral response can be achieved upon administration of the prodrug.


Cancer Chemotherapy and Pharmacology | 2009

Delivery of high levels of anti-proliferative nucleoside triphosphates to CYP3A-expressing cells as a potential treatment for hepatocellular carcinoma

Deidre A. MacKenna; Annika C. Montag; Serge H. Boyer; David L. Linemeyer; Mark D. Erion

PurposeHepatocellular carcinoma (HCC) is a life-threatening condition with only one drug treatment regimen approved for use. Oncolytic nucleosides are minimally effective against HCC putatively because of their inability to achieve cytotoxic levels of the active metabolite [nucleoside triphosphate (NTP)] in tumor cells at doses that are well tolerated. The aim of our studies was to explore the utility of CYP3A-activated prodrugs of cytarabine and fludarabine monophosphate for the treatment of HCC.MethodsProdrugs of cytarabine and fludarabine monophosphates were evaluated for their ability to safely achieve NTP levels in the liver of normal mice that are cytotoxic to hepatoma cells.ResultsWhile therapeutic levels of NTPs are achieved in the livers of normal rodents after administration of the prodrugs, only MB07133 achieved these levels whithout exhibiting signs of liver toxicity or myelosuppression.ConclusionsAs the levels of araCTP achieved in the liver at therapeutic doses are only toxic to proliferating cells (such as those in HCC tumors), but not the non-proliferative adjacent tissue, MB07133 treatment has the potential to be both efficacious and well tolerated in HCC patients.


Journal of Medicinal Chemistry | 2007

Liver-Targeted Prodrugs of 2‘-C-Methyladenosine for Therapy of Hepatitis C Virus Infection

Scott J. Hecker; K. Raja Reddy; Paul D. van Poelje; Zhili Sun; Wenjian Huang; Vaibhav Varkhedkar; M. Venkat Reddy; James M. Fujitaki; David B. Olsen; Kenneth A. Koeplinger; Serge H. Boyer; David L. Linemeyer; Malcolm Maccoss; Mark D. Erion


Hepatology | 2003

524 MB07133: a hepdirect™ prodrug of cytarabine monophosphate for use in hepatocellular carcinoma

Deidre A. MacKenna; Serge H. Boyer; Annika C. Montag; Barbara Treash-Osio; Paul A. Rolzin; David L. Linemeyer; Mark D. Erion

Collaboration


Dive into the David L. Linemeyer's collaboration.

Top Co-Authors

Avatar

Mark D. Erion

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bruce R. Ito

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edward E. Cable

Penn State Milton S. Hershey Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kenneth A. Koeplinger

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge