Co-reporter:Brian R. Blank, Jiri Gut, Philip J. Rosenthal, and Adam R. Renslo
Journal of Medicinal Chemistry July 27, 2017 Volume 60(Issue 14) pp:6400-6400
Publication Date(Web):July 10, 2017
DOI:10.1021/acs.jmedchem.7b00699
We describe the first systematic study of antimalarial 1,2,4-trioxolanes bearing a substitution pattern regioisomeric to that of arterolane. Conformational analysis suggested that trans-3″-substituted trioxolanes would exhibit Fe(II) reactivity and antiparasitic activity similar to that achieved with canonical cis-4″ substitution. The chiral 3″ analogues were prepared as single stereoisomers and evaluated alongside their 4″ congeners against cultured malaria parasites and in a murine malaria model. As predicted, the trans-3″ analogues exhibited in vitro antiplasmodial activity remarkably similar to that of their cis-4″ comparators. In contrast, efficacy in the Plasmodium berghei mouse model differed dramatically for some of the congeneric pairs. The best of the novel 3″ analogues (e.g., 12i) outperformed arterolane itself, producing cures in mice after a single oral exposure. Overall, this study suggests new avenues for modulating Fe(II) reactivity and the pharmacokinetic and pharmacodynamic properties of 1,2,4-trioxolane antimalarials.
Co-reporter:Benjamin Spangler, Shaun D. Fontaine, Yihui Shi, Lidia Sambucetti, Aras N. Mattis, Byron Hann, James A. Wells, and Adam R. Renslo
Journal of Medicinal Chemistry 2016 Volume 59(Issue 24) pp:11161-11170
Publication Date(Web):November 21, 2016
DOI:10.1021/acs.jmedchem.6b01470
Here we describe a new approach for tumor targeting in which augmented concentrations of Fe(II) in cancer cells and/or the tumor microenvironment triggers drug release from an Fe(II)-reactive prodrug conjugate. The 1,2,4-trioxolane scaffold developed to enable this approach can in principle be applied to a broad range of cancer therapeutics and is illustrated here with Fe(II)-targeted forms of a microtubule toxin and a duocarmycin-class DNA-alkylating agent. We show that the intrinsic reactivity/toxicity of the duocarmycin analog is masked in the conjugated form and this greatly reduced toxicity in mice. This in turn permitted elevated dosing levels, leading to higher systemic exposure and a significantly improved response in tumor xenograft models. Overall our results suggest that Fe(II)-dependent drug delivery via trioxolane conjugates could have significant utility in expanding the therapeutic index of a range of clinical and preclinical stage cancer chemotherapeutics.
Co-reporter:Erica M. W. Lauterwasser, Shaun D. Fontaine, Hao Li, Jiri Gut, Kasiram Katneni, Susan A. Charman, Philip J. Rosenthal, Matthew Bogyo, and Adam R. Renslo
ACS Medicinal Chemistry Letters 2015 Volume 6(Issue 11) pp:1145
Publication Date(Web):October 2, 2015
DOI:10.1021/acsmedchemlett.5b00296
Peroxidic antimalarial agents including the sequiterpene artemisinins and the synthetic 1,2,4-trioxolanes function via initial intraparasitic reduction of an endoperoxide bond. By chemically coupling this reduction to release of a tethered drug species it is possible to confer two distinct pharmacological effects in a parasite-selective fashion, both in vitro and in vivo. Here we demonstrate the trioxolane-mediated delivery of the antimalarial agent mefloquine in a mouse malaria model. Selective partitioning of the trioxolane–mefloquine conjugate in parasitized erythrocytes, combined with effective exclusion of the conjugate from brain significantly reduced brain exposure as compared to mice directly administered mefloquine. These studies suggest the potential of trioxolane-mediated drug delivery to mitigate off-target effects of existing drugs, including the adverse neuropsychiatric effects of mefloquine use in therapeutic and chemoprophylactic settings.Keywords: antimalarial; drug delivery; mefloquine; trioxolane
Co-reporter:R. Jeffrey Neitz, Clifford Bryant, Steven Chen, Jiri Gut, Estefania Hugo Caselli, Servando Ponce, Somenath Chowdhury, Haichao Xu, Michelle R. Arkin, Jonathan A. Ellman, Adam R. Renslo
Bioorganic & Medicinal Chemistry Letters 2015 Volume 25(Issue 21) pp:4834-4837
Publication Date(Web):1 November 2015
DOI:10.1016/j.bmcl.2015.06.066
Inhibition of the cysteine protease cruzain from Trypanosoma cruzi has been studied pre-clinically as a new chemotherapeutic approach to treat Chagas’ disease. Efficacious effects of vinylsulfone-based cruzain inhibitors in animal models support this therapeutic hypothesis. More recently, substrate–activity screening was used to identify nonpeptidic tetrafluorophenoxymethyl ketone inhibitors of cruzain that showed promising efficacy in animal models. Herein we report efforts to further optimize the in vitro potency and in vivo pharmacokinetic properties of this new class of cruzain inhibitors. Through modifications of the P1, P2 and/or P3 positions, new analogs have been identified with reduced lipophilicity, enhanced potency, and improved oral exposure and bioavailability.
Co-reporter:Dr. Shaun D. Fontaine;Benjamin Spangler;Dr. Jiri Gut;Dr. Erica M. W. Lauterwasser;Dr. Philip J. Rosenthal;Dr. Adam R. Renslo
ChemMedChem 2015 Volume 10( Issue 1) pp:47-51
Publication Date(Web):
DOI:10.1002/cmdc.201402362
Abstract
Antimalarial agents artemisinin and arterolane act via initial reduction of a peroxide bond in a process likely mediated by ferrous iron sources in the parasite. Here, we report the synthesis and antiplasmodial activity of arterolane-like 1,2,4-trioxolanes specifically designed to release a tethered drug species within the malaria parasite. Compared with our earlier drug delivery scaffolds, these new arterolane-inspired systems are of significantly decreased molecular weight and possess superior metabolic stability. We describe an efficient, concise and scalable synthesis of the new systems, and demonstrate the use of the aminonucleoside antibiotic puromycin as a chemo/biomarker to validate successful drug release in live Plasmodium falciparum parasites. Together, the improved drug-like properties, more efficient synthesis, and proof of concept using puromycin, suggests these new molecules as improved vehicles for targeted drug delivery to the malaria parasite.
Co-reporter:Shaun D. Fontaine, Antonio G. DiPasquale, and Adam R. Renslo
Organic Letters 2014 Volume 16(Issue 21) pp:5776-5779
Publication Date(Web):October 21, 2014
DOI:10.1021/ol5028392
Ferrous iron-promoted reduction of a hindered peroxide bond underlies the antimalarial action of the 1,2,4-trioxane artemisinin and the 1,2,4-trioxolane arterolane. In appropriately designed systems, a 1,2,4-trioxolane ring can serve as a trigger to realize ferrous iron-dependent and parasite-selective drug delivery, both in vitro and in vivo. A stereocontrolled, expeditious (three steps), and efficient (67–71% overall yield) synthesis of 1,2,4-trioxolanes possessing the requisite 3″ substitution pattern that enables ferrous iron-dependent drug delivery is reported. The key synthetic step involves a diastereoselective Griesbaum co-ozonolysis reaction to afford primarily products with a trans relationship between the 3″ substituent and the peroxide bridge, as confirmed by X-ray structural analysis of a 3″-substituted 4-nitrobenzoate analogue.
Co-reporter:Sina Ghaemmaghami ; Miranda Russo
Journal of Medicinal Chemistry 2014 Volume 57(Issue 16) pp:6919-6929
Publication Date(Web):April 24, 2014
DOI:10.1021/jm5001425
Creutzfeldt–Jakob disease (CJD) is a rare but invariably fatal neurodegenerative disease caused by misfolding of an endogenous protein into an alternative pathogenic conformation. The details of protein misfolding and aggregation are not well understood nor are the mechanism(s) by which the aggregated protein confers cellular toxicity. While there is as yet no clear consensus about how best to intervene therapeutically in CJD, prion infections can be propagated in cell culture and in experimental animals, affording both in vitro and in vivo models of disease. Here we review recent lead discovery efforts for CJD, with a focus on our own efforts to optimize 2-aminothiazole analogues for anti-prion potency in cells and for brain exposure in mice. The compounds that emerged from this effort were found to be efficacious in multiple animal models of prion disease even as they revealed new challenges for the field, including the emergence of resistant prion strains.
Co-reporter:David M. Turner, Christopher T. M. B. Tom, and Adam R. Renslo
ACS Combinatorial Science 2014 Volume 16(Issue 12) pp:661
Publication Date(Web):October 29, 2014
DOI:10.1021/co500132q
Disulfide exchange screening is a site-directed approach to fragment-based lead discovery that requires a bespoke library of disulfide-containing fragments. Previously, we described a simple one-pot, two-step synthesis of disulfide fragments from amine- or acid-bearing starting materials. Here, we describe the synthesis of disulfide fragments that bear a 1,4-substituted-1,2,3-triazole linkage between disulfide and molecular diversity element. This work establishes the compatibility of copper(I)-catalyzed azide–alkyne cycloaddition (CuAAC) chemistry with a one-pot, two-step reaction sequence that can be readily parallelized. We performed 96 reactions in a single deep-well microtiter plate, employing 48 alkynes and two different azide linker reagents. From this effort, a total of 81 triazole-containing disulfide fragments were obtained in useful isolated yields. Thus, CuAAC chemistry offers an experimentally convenient method to rapidly prepare disulfide fragments that are structurally distinct from fragments accessed via amide, sulfonamide, or isocyanate chemistries.Keywords: click chemistry; disulfide exchange screening; disulfide synthesis; tethering
Co-reporter:Dr. Jeremy Murray;Dr. Anthony M. Giannetti;Micah Steffek;Paul Gibbons;Dr. Brian R. Hearn;Dr. Frederick Cohen;Christine Tam;Christine Pozniak;Bron Bravo;Dr. Joe Lewcock;Priyadarshini Jaishankar;Cuong Q. Ly;Xianrui Zhao;Dr. Yinyan Tang;Dr. Preeti Chugha; Michelle R. Arkin;Dr. John Flygare; Adam R. Renslo
ChemMedChem 2014 Volume 9( Issue 1) pp:
Publication Date(Web):
DOI:10.1002/cmdc.201390058
Co-reporter:Dr. Jeremy Murray;Dr. Anthony M. Giannetti;Micah Steffek;Paul Gibbons;Dr. Brian R. Hearn;Dr. Frederick Cohen;Christine Tam;Christine Pozniak;Bron Bravo;Dr. Joe Lewcock;Priyadarshini Jaishankar;Cuong Q. Ly;Xianrui Zhao;Dr. Yinyan Tang;Dr. Preeti Chugha; Michelle R. Arkin;Dr. John Flygare; Adam R. Renslo
ChemMedChem 2014 Volume 9( Issue 1) pp:73-77
Publication Date(Web):
DOI:10.1002/cmdc.201300424
Abstract
Although they represent attractive therapeutic targets, caspases have so far proven recalcitrant to the development of drugs targeting the active site. Allosteric modulation of caspase activity is an alternate strategy that potentially avoids the need for anionic and electrophilic functionality present in most active-site inhibitors. Caspase-6 has been implicated in neurodegenerative disease, including Huntington’s and Alzheimer’s diseases. Herein we describe a fragment-based lead discovery effort focused on caspase-6 in its active and zymogen forms. Fragments were identified for procaspase-6 using surface plasmon resonance methods and subsequently shown by X-ray crystallography to bind a putative allosteric site at the dimer interface. A fragment-merging strategy was employed to produce nanomolar-affinity ligands that contact residues in the L2 loop at the dimer interface, significantly stabilizing procaspase-6. Because rearrangement of the L2 loop is required for caspase-6 activation, our results suggest a strategy for the allosteric control of caspase activation with drug-like small molecules.
Co-reporter:Adam R. Renslo
ACS Medicinal Chemistry Letters 2013 Volume 4(Issue 12) pp:1126-1128
Publication Date(Web):November 13, 2013
DOI:10.1021/ml4004414
The search for antimalarial remedies predates modern medicine and the concept of small molecule chemotherapy, yet has played a central role in the development of both. This history is reviewed in the context of the current renaissance in antimalarial drug discovery, which is seeing modern drug discovery approaches applied to the problem for the first time. Great strides have been made in the past decade, but further innovations from the drug discovery community will be required if the ultimate dream of eradication is to be achieved.
Co-reporter:Edgar Deu;Ingrid T. Chen;Erica M. W. Lauterwasser;Juan Valderramos;Hao Li;Laura E. Edgington;Matthew Bogyo
PNAS 2013 Volume 110 (Issue 45 ) pp:18244-18249
Publication Date(Web):2013-11-05
DOI:10.1073/pnas.1312782110
The precise targeting of cytotoxic agents to specific cell types or cellular compartments is of significant interest in medicine,
with particular relevance for infectious diseases and cancer. Here, we describe a method to exploit aberrant levels of mobile
ferrous iron (FeII) for selective drug delivery in vivo. This approach makes use of a 1,2,4-trioxolane moiety, which serves as an FeII-sensitive “trigger,” making drug release contingent on FeII-promoted trioxolane fragmentation. We demonstrate in vivo validation of this approach with the Plasmodium berghei model of murine malaria. Malaria parasites produce high concentrations of mobile ferrous iron as a consequence of their catabolism
of host hemoglobin in the infected erythrocyte. Using activity-based probes, we successfully demonstrate the FeII-dependent and parasite-selective delivery of a potent dipeptidyl aminopeptidase inhibitor. We find that delivery of the compound
in its FeII-targeted form leads to more sustained target inhibition with greatly reduced off-target inhibition of mammalian cathepsins.
This selective drug delivery translates into improved efficacy and tolerability. These findings demonstrate the utility of
a purely chemical means to achieve selective drug targeting in vivo. This approach may find useful application in parasitic
infections and more broadly in any disease state characterized by aberrant production of reactive ferrous iron.
Co-reporter:Salma B. Rafi ; Brian R. Hearn ; Punitha Vedantham ; Matthew P. Jacobson
Journal of Medicinal Chemistry 2012 Volume 55(Issue 7) pp:3163-3169
Publication Date(Web):March 6, 2012
DOI:10.1021/jm201634q
We evaluate experimentally and computationally the membrane permeability of matched sets of peptidic small molecules bearing natural or bioisosteric unnatural amino acids. We find that the intentional introduction of hydrogen bond acceptor–donor pairs in such molecules can improve membrane permeability while retaining or improving other favorable drug-like properties. We employ an all-atom force field based method to calculate changes in free energy associated with the transfer of the peptidic molecules from water to membrane. This computational method correctly predicts rank order experimental permeability trends within congeneric series and is much more predictive than calculations (e.g., clogP) that do not consider three-dimensional conformation.
Co-reporter:Derek A. Nichols ; Priyadarshini Jaishankar ; Wayne Larson ; Emmanuel Smith ; Guoqing Liu ; Racha Beyrouthy ; Richard Bonnet ; Adam R. Renslo ;Yu Chen
Journal of Medicinal Chemistry 2012 Volume 55(Issue 5) pp:2163-2172
Publication Date(Web):February 1, 2012
DOI:10.1021/jm2014138
The emergence of CTX-M class A extended-spectrum β-lactamases poses a serious health threat to the public. We have applied structure-based design to improve the potency of a novel noncovalent tetrazole-containing CTX-M inhibitor (Ki = 21 μM) more than 200-fold via structural modifications targeting two binding hot spots, a hydrophobic shelf formed by Pro167 and a polar site anchored by Asp240. Functional groups contacting each binding hot spot independently in initial designs were later combined to produce analogues with submicromolar potencies, including 6-trifluoromethyl-3H-benzoimidazole-4-carboxylic acid [3-(1H-tetrazol-5-yl)-phenyl]-amide, which had a Ki value of 89 nM and reduced the MIC of cefotaxime by 64-fold in CTX-M-9 expressing Escherichia coli. The in vitro potency gains were accompanied by improvements in ligand efficiency (from 0.30 to 0.39) and LipE (from 1.37 to 3.86). These new analogues represent the first nM-affinity noncovalent inhibitors of a class A β-lactamase. Their complex crystal structures provide valuable information about ligand binding for future inhibitor design.
Co-reporter:Carmony L. Hartwig ; Erica M.W. Lauterwasser ; Sumit S. Mahajan ; Jonathan M. Hoke ; Roland A. Cooper
Journal of Medicinal Chemistry 2011 Volume 54(Issue 23) pp:8207-8213
Publication Date(Web):October 24, 2011
DOI:10.1021/jm2012003
The 1,2,4-trioxolanes are a new class of synthetic peroxidic antimalarials currently in human clinical trials. The well-known reactivity of the 1,2,4-trioxolane ring toward inorganic ferrous iron and ferrous iron heme is proposed to play a role in the antimalarial action of this class of compounds. We have designed structurally relevant fluorescent chemical probes to study the subcellular localization of 1,2,4-trioxolanes in cultured Plasmodium falciparum parasites. Microscopy experiments revealed that a probe fluorescently labeled on the adamantane ring accumulated specifically in digestive vacuole-associated neutral lipid bodies within the parasite while an isosteric, but nonperoxidic, congener did not. Probes fluorescently labeled on the cyclohexane ring showed no distinct localization pattern. In their subcellular localization and peroxidative effects, 1,2,4-trioxolane probes behave much like artemisinin-based probes studied previously. Our results are consistent with a role for adamantane-derived carbon-centered radicals in the antimalarial action of 1,2,4-trioxolanes, as hypothesized previously on the basis of chemical reactivity studies.
Co-reporter:Alejandra Gallardo-Godoy ; Joel Gever ; Kimberly L. Fife ; B. Michael Silber ; Stanley B. Prusiner
Journal of Medicinal Chemistry 2011 Volume 54(Issue 4) pp:1010-1021
Publication Date(Web):January 19, 2011
DOI:10.1021/jm101250y
2-Aminothiazoles are a new class of small molecules with antiprion activity in prion-infected neuroblastoma cell lines ( J. Virol. 2010, 84, 3408). We report here structure−activity studies undertaken to improve the potency and physiochemical properties of 2-aminothiazoles, with a particular emphasis on achieving and sustaining high drug concentrations in the brain. The results of this effort include the generation of informative structure−activity relationships (SAR) and the identification of lead compounds that are orally absorbed and achieve high brain concentrations in animals. The new aminothiazole analogue (5-methylpyridin-2-yl)-[4-(3-phenylisoxazol-5-yl)-thiazol-2-yl]-amine (27), for example, exhibited an EC50 of 0.94 μM in prion-infected neuroblastoma cells (ScN2a-cl3) and reached a concentration of ∼25 μM in the brains of mice following three days of oral administration in a rodent liquid diet. The studies described herein suggest 2-aminothiazoles as promising new leads in the search for effective therapeutics for prion diseases.
Co-reporter:Dr. Sumit S. Mahajan;Dr. Edgar Deu;Dr. Erica M. W. Lauterwasser;Melissa J. Leyva;Dr. Jonathan A. Ellman;Dr. Matthew Bogyo;Dr. Adam R. Renslo
ChemMedChem 2011 Volume 6( Issue 3) pp:415-419
Publication Date(Web):
DOI:10.1002/cmdc.201100002
Co-reporter:Dr. Sumit S. Mahajan;Dr. Edgar Deu;Dr. Erica M. W. Lauterwasser;Melissa J. Leyva;Dr. Jonathan A. Ellman;Dr. Matthew Bogyo;Dr. Adam R. Renslo
ChemMedChem 2011 Volume 6( Issue 3) pp:
Publication Date(Web):
DOI:10.1002/cmdc.201190006
Co-reporter:Rafaela S. Ferreira ; Clifford Bryant ; Kenny K. H. Ang ; James H. McKerrow ; Brian K. Shoichet
Journal of Medicinal Chemistry 2009 Volume 52(Issue 16) pp:5005-5008
Publication Date(Web):July 28, 2009
DOI:10.1021/jm9009229
A docking screen identified reversible, noncovalent inhibitors (e.g., 1) of the parasite cysteine protease cruzain. Chemical optimization of 1 led to a series of oxadiazoles possessing interpretable SAR and potencies as much as 500-fold greater than 1. Detailed investigation of the SAR series subsequently revealed that many members of the oxadiazole class (and surprisingly also 1) act via divergent modes of inhibition (competitive or via colloidal aggregation) depending on the assay conditions employed.
Co-reporter:Clifford Bryant, Iain D. Kerr, Moumita Debnath, Kenny K.H. Ang, Joseline Ratnam, Rafaela S. Ferreira, Priyadarshini Jaishankar, DongMei Zhao, Michelle R. Arkin, James H. McKerrow, Linda S. Brinen, Adam R. Renslo
Bioorganic & Medicinal Chemistry Letters 2009 Volume 19(Issue 21) pp:6218-6221
Publication Date(Web):1 November 2009
DOI:10.1016/j.bmcl.2009.08.098
We describe here the identification of non-peptidic vinylsulfones that inhibit parasite cysteine proteases in vitro and inhibit the growth of Trypanosoma brucei brucei parasites in culture. A high resolution (1.75 Å) co-crystal structure of 8a bound to cruzain reveals how the non-peptidic P2/P3 moiety in such analogs bind the S2 and S3 subsites of the protease, effectively recapitulating important binding interactions present in more traditional peptide-based protease inhibitors and natural substrates.We describe here the identification of non-peptidic vinylsulfones that inhibit parasite cysteine proteases in vitro and inhibit the growth of Trypanosoma brucei brucei parasites in culture. A high resolution (1.75 Å) co-crystal structure of 8a bound to cruzain reveals how the non-peptidic P2/P3 moiety in such analogs bind the S2 and S3 subsites of the protease, effectively recapitulating important binding interactions present in more traditional peptide-based protease inhibitors and natural substrates.
Co-reporter:Adam R. Renslo, Gary W. Luehr, Stuart Lam, Neil E. Westlund, Marcela Gómez, Corrine J. Hackbarth, Dinesh V. Patel, Mikhail F. Gordeev
Bioorganic & Medicinal Chemistry Letters 2006 Volume 16(Issue 13) pp:3475-3478
Publication Date(Web):1 July 2006
DOI:10.1016/j.bmcl.2006.03.104
A new series of antimicrobial oxazolidinones bearing unsaturated heterocyclic C-rings is described. Dihydrothiopyran derivatives were prepared from the saturated tetrahydrothiopyran sulfoxides via a Pummerer-rearrangement/elimination sequence. Two new synthetic approaches to the dihydrothiazine ring system were explored, the first involving a novel trifluoroacetylative-detrifluoroacetylative Pummerer-type reaction sequence and the second involving direct dehydrogenation of tetrahydrothiopyran S,S-dioxide intermediates. Final analogs such as 4 and 13 represent oxidized congeners of recent pre-clinical and clinical oxazolidinones.A new series of oxazolidinone analogs bearing unsaturated sulfur-containing C-rings is described. New synthetic approaches to the dihydrothiazine ring system are also disclosed.