NIH Public Access Author Manuscript Bioorg Med Chem Lett. Author manuscript; available in PMC 2013 June 15.

Similar documents
ARV Mode of Action. Mode of Action. Mode of Action NRTI. Immunopaedia.org.za

Received 29 May 2003/Returned for modification 7 August 2003/Accepted 13 November 2003

In vitro Suppression of K65R Reverse Transcriptase-mediated Tenofovirand. Adefovir-5' Diphosphate Resistance Conferred by the Borano- ACCEPTED

Antiviral Activity of Tenofovir Alafenamide against HIV-1 with Thymidine Analog Mutation(s) and M184V

PREDICTING SUSCEPTIBILITY AND RESISTANCE FOR HEPATITIS B VIRUS CAPSID ASSEMBLY EFFECTORS

Antiviral Agents I. Tutorial 6

The E138A substitution in HIV-1 reverse transcriptase decreases in vitro. susceptibility to emtricitabine as indicated by competitive fitness assays

7.014 Problem Set 7 Solutions

L I F E S C I E N C E S

Les thérapeutiques antirétrovirales : entre nouveauté et expérience

Molecular Mechanism by which K70E in HIV-1 Reverse. Transcriptase Confers Resistance to Nucleoside Reverse ACCEPTED

Human Genome: Mapping, Sequencing Techniques, Diseases

Structural Aspects of Drug Resistance and Inhibition of HIV-1 Reverse Transcriptase

Patrick: An Introduction to Medicinal Chemistry 5e Chapter 09

Why Do HIV-1 and HIV-2 Use Different Pathways to Develop AZT Resistance?

October 26, Lecture Readings. Vesicular Trafficking, Secretory Pathway, HIV Assembly and Exit from Cell

WHY IS THIS IMPORTANT?

Section 6. Junaid Malek, M.D.

Nicolas Sluis-Cremer,* Chih-Wei Sheen, Shannon Zelina, Pedro S. Argoti Torres, Urvi M. Parikh, and John W. Mellors

Zalcitabine 2-3 -dideoxycytidine, ddc TUD DaMocles SoSe 15

Management of NRTI Resistance

HIV replication and selection of resistance: basic principles

HIV - Life cycle. HIV Life Cyle

Supplementary Figure 1. SC35M polymerase activity in the presence of Bat or SC35M NP encoded from the phw2000 rescue plasmid.

Antiviral Therapy 6:

Molecular Dynamics of HIV-1 Reverse Transcriptase

Antiviral Agents DEPARTEMEN FARMAKOLOGI & TERAPEUTIK FK USU. 06 August

Jiong Wang, 1 Dongge Li, 1 Robert A. Bambara, 2 Hongmei Yang 3 and Carrie Dykes 1 INTRODUCTION

Patrick: An Introduction to Medicinal Chemistry 5e Chapter 06

Hepatitis B Antiviral Drug Development Multi-Marker Screening Assay

Emerging CMV Resistance Profile for CMX001

Vemlidy. (tenofovir alafenamide) New Product Slideshow

Tenofovir disoproxil STADA 245 mg film-coated tablets , Version 1.2 PUBLIC SUMMARY OF THE RISK MANAGEMENT PLAN

Biology 5A Fall 2010 Macromolecules Chapter 5

Steps in viral replication (I)

Betancor et al. Retrovirology 2012, 9:68

Effects of the Translocation Status of Human Immunodeficiency Virus Type 1 Reverse Transcriptase on the Efficiency of Excision of Tenofovir

Antiviral Chemotherapy

2.2 Cell Construction

Descovy. (emtricitabine, tenofovir alafenamide) New Product Slideshow

Title: Sofosbuvir: an anti-viral drug with potential efficacy against Zika infection

Mechanistic Differences in RNA-dependent DNA Polymerization and Fidelity between Murine Leukemia Virus and HIV-1 Reverse Transcriptases*

Supplementary information

Sofosbuvir (Sovaldi) Drug Summary. Class and Mechanism. Table of Contents

Forward Looking Statements

HEPATITIS B: are escape mutants of concern?

Evaluation and Management of Virologic Failure

Molecular Biology (BIOL 4320) Exam #2 May 3, 2004

PRINCIPLES and TRENDS in MANAGEMENT of HIV DISEASE: PROBLEMS OF DRUG RESISTANCE in VIRUSES of DIFFERENT SUBTYPES

Molecular building blocks

Antivirals. Lecture 20 Biology 3310/4310 Virology Spring 2017

Enzymes Part III: regulation II. Dr. Mamoun Ahram Summer, 2017

modified dye uptake assay including formazan test EC 90 not tested plaque reduction assay

It has been estimated that 90% of individuals

Life Sciences 1A Midterm Exam 2. November 13, 2006

-are poly-hydroxylated aldehydes and ketones -can cyclise -can form polymeric chains

HIV 101: Fundamentals of HIV Infection

NEXT GENERATION SEQUENCING OPENS NEW VIEWS ON VIRUS EVOLUTION AND EPIDEMIOLOGY. 16th International WAVLD symposium, 10th OIE Seminar

Treatment of respiratory virus infection Influenza A & B Respiratory Syncytial Virus (RSV)

Virus Genetic Diversity

Journal of Microbes and Infection,June 2007,Vol 2,No. 2. (HBsAg)2 , (PCR) 1762/ 1764

Basics of hepatitis B diagnostics. Dr Emma Page MRCP MD(Res) Locum Consultant Sexual Health & Virology

CLINICAL PEARLS OF NEW HIV MEDICATIONS PHARMACIST OBJECTIVES TECHNICIAN OBJECTIVES. At the end of this presentation pharmacists will be able to:

Medicinal Chemistry. Antiviral Agents

Antiviral Chemotherapy

RNA (Ribonucleic acid)

*viruses have no cell wall and made up of nucleic acid components.

LS1a Fall 2014 Problem Set #4 Due Monday 11/3 at 6 pm in the drop boxes on the Science Center 2 nd Floor

Antiviral Drugs. Munir Gharaibeh MD, PhD, MHPE School of Medicine, The University of Jordan November 2018

172R 172K TAM-2/172R TAM-2/172K. AZT concentration [nm] AZT concentration [nm] MgCl 2 2.5K 2.5K 5K 2.5K 5K 2.5K K 5K 2.5K 5K 2.5K 50 2.

LESSON 4.4 WORKBOOK. How viruses make us sick: Viral Replication

Antibacterials and Antivirals

Evidence Review: Comparison between tenofovir alafenamide and tenofovir disoproxil fumarate. February For public consultation

Differentiating emtricitabine (FTC) from lamivudine (3TC): what a fine-tuning of antiretroviral therapy might entail

Supplementary Figure 1. FACS analysis of cells infected with TY93/H5N1 GFP-627E,

Antiviral Drugs. Munir Gharaibeh MD, PhD, MHPE School of Medicine, The University of Jordan November 2018

Resistance profile of the new nucleoside reverse transcriptase inhibitor apricitabine

Reverse transcription and integration

Short polymer. Dehydration removes a water molecule, forming a new bond. Longer polymer (a) Dehydration reaction in the synthesis of a polymer

Previous Class. Today. Detection of enzymatic intermediates: Protein tyrosine phosphatase mechanism. Protein Kinase Catalytic Properties

L I F E S C I E N C E S

7.012 Quiz 3 Answers

Alan Weiner BIOCHEM 530 Friday, MEB 248 October 23, 2015 RNA structure, the ribosome, structure-based drug design

Diagnostic Methods of HBV and HDV infections

KEY NAME (printed very legibly) UT-EID

Monitoring for Drug Resistance by Genotyping. Urvi M Parikh, PhD MTN Virology Core Lab

Acute Hepatitis B Virus Infection with Recovery

Molecular Mechanisms by Which Human Immunodeficiency Virus Type 1 Integrase Stimulates the Early Steps of Reverse Transcription

Life Science 1A Final Exam. January 19, 2006

AP Biology Summer Assignment Chapter 3 Quiz

CDC website:

Statin inhibition of HMG-CoA reductase: a 3-dimensional view

Received 19 May 2004/Returned for modification 3 August 2004/Accepted 8 September 2004

HIV-1 infection has been the target of various multidrug therapies,

The building blocks of life.

The Carbon Atom (cont.)

Supplementary information. MARCH8 inhibits HIV-1 infection by reducing virion incorporation of envelope glycoproteins

X/01/$ DOI: /JVI Copyright 2001, American Society for Microbiology. All Rights Reserved.

HSV DNA replication. Herpesvirus Latency. Latency and Chemotherapy. Human Herpesviruses - subtypes. Acyclovir (acycloguanosine) {Zovirax}

Chemistry 2050 Introduction to Organic Chemistry Fall Semester 2011 Dr. Rainer Glaser

Transcription:

NIH Public Access Author Manuscript Published in final edited form as: Bioorg Med Chem Lett. 2012 June 15; 22(12): 4064 4067. doi:10.1016/j.bmcl.2012.04.078. Pre-steady state kinetic analysis of cyclobutyl derivatives of 2 - deoxyadenosine 5 -triphosphate as inhibitors of HIV-1 reverse transcriptase Jiae Kim a, Ligong Wang a,1, Yongfeng Li b, Kimberlynne D. Becnel b, Kathleen M. Frey a, Scott J. Garforth c,2, Vinayaka R. Prasad c, Raymond F. Schinazi d, Dennis C. Liotta b, and Karen S. Anderson a,* a Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520, USA b Department of Chemistry, Emory University, Atlanta, GA 30322, USA c Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA d Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Veterans Affairs Medical Center, Atlanta, GA 30322, USA Abstract Pre-steady state kinetic analysis was utilized for biochemical evaluation of a series of cyclobutyl adenosine nucleotide analogs with HIV-1 RT WT. The phosphonyl-diphosphate form of the cyclobutyl nucleotide, 5, was the most efficiently incorporated of the series. Nucleotide 5 was 4- fold more efficiently incorporated than the FDA approved TFV-DP by RT WT. The kinetics of incorporation for 5 using the drug resistant mutant enzyme K65R was also determined. 5 was 3- fold more efficiently incorporated compared to TFV-DP with RT K65R. These results demonstrate cyclobutyl adenosine analogs can act as substrates for incorporation by HIV-1 RT and be a potential scaffold for HIV inhibitors. Keywords HIV-1 RT; Cyclobutyl adenosine analogs; tenofovir; K65R mutant of RT; pre-steady state kinetics Human immunodeficiency virus (HIV) affects millions of people around the world. Currently, therapeutics are available for the treatment of HIV-1 with nucleoside analogs 2012 Elsevier Ltd. All rights reserved. * To whom correspondence should be addressed: Yale University School of Medicine, Dept. of Pharmacology, SHM B350B, 333 Cedar Street, New Haven, CT 06520-8066. Tel.: 203-785-4526; Fax: 203-785-7670; karen.anderson@yale.edu. 1 Current Address: GE Healthcare, 800 Centennial Ave. Piscataway, NJ 08854 2 Current Address: Macromolecular Therapeutics Development Facility, Albert Einstein College of Medicine, Bronx, NY 10461, USA Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. Supporting Information This file contains information regarding the synthesis for compounds 3, 5, and 6; information on the docking model created of compound 5 in the HIV-1 RT active site; and Supp. Figure 1 which indicates the ability of compound 5 to act as a chain terminator after incorporation by HIV-1 RT.

Kim et al. Page 2 continuing to be the backbone for the majority of persons infected. This class of drugs, in their triphosphate form target the enzyme reverse transcriptase (RT). This polymerase is essential for viral replication and is responsible for the conversion of viral RNA into the double stranded proviral DNA which will be subsequently be integrated into the host cell s genome. One class of therapeutics available is the nucleoside reverse transcriptase inhibitors (NRTIs). These nucleoside analogs, which are phosphorylated to the triphosphate form by cellular kinases act as a substrate for HIV-1 RT and are incorporated into the growing strand of viral DNA. Due to the lack of the 3'-hydroxyl group, these analogs act as chain terminators and do not allow for further elongation of the viral DNA. Although there are many NRTIs available, there is still a need for novel and more potent inhibitors. Clinical use of FDA approved NRTIs give rise to drug resistant mutations in RT that render these inhibitors ineffective. One such mutation is K65R, which causes resistance against tenofovir (TFV or PMPA). 1, 2 The emergence of such drug resistant viruses gives the need for new NRTIs with different scaffolds to be explored as potential therapeutics. Successful cyclobutyl nucleosides have been developed as antiviral agents, such as Lobucavir against HBV. A cyclobutyl analog of cytosine triphosphate was found to be effective against WT and the M184V drug resistant form of HIV-1 RT. 3 In this paper, we report the ability of cyclobutyl adenosine derivatives to be incorporated by HIV-1 RT. In order to examine the analogs ability to act as a substrate, we utilized pre-steady state kinetics. With the use of a DNA/DNA primer-template containing the primer binding site (PBS) and its complementary sequence, we examined the kinetics to determine the incorporation efficiency for the natural substrate, datp, as well as the analogs. Three different compounds were synthesized in this series cyclobutyl-adenosine triphosphate 3, 2'-fluoro-cyclobutyl adenosine triphosphate 4, and cyclobutyl-adenosinephosphonyldiphosphate 5. I Details on the synthesis have been reported separately, a brief synopsis of the synthesis is included in the Supporting Information. These compounds, along with the natural substrate 2 -deoxyadenosine 5 -triphosphate (datp) 1, and the acyclic FDA approved 2 -deoxyadenosine nucleotide tenofovir diphosphate (TFV-DP) 2, were tested for their ability to elongate a radiolabeled primer-template under the conditions of 250 nm HIV-1 RT (active site concentration), 50 nm primer-template, in the presence of 10 mm MgCl 2, and various concentrations of the nucleotides as previously described. 4 The natural substrate was most efficiently incorporated, compared to the derivatives as shown in Table 1. Tenofovir diphosphate 2, was 56-fold less efficient for incorporation compared to the natural substrate. This decrease in efficiency was largely due to the 15-fold decrease in the maximum rate of incorporation (k pol ) and a 4-fold decrease in the binding affinity of the nucleotide as observed by the increase in the binding affinity constant (K d ). Previous kinetic work has been performed with TFV-DP 2 and demonstrates a decrease in incorporation efficiency of the analog versus datp with varying degrees, which may be due to the use of different primer-template sequences. 1, 5, 6 The presence of a cyclobutyl ring, 3, decreased the incorporation efficiency by over 200-fold (2.7 s 1 µm 1 for 1 versus 0.011 s 1 µm 1 for 3), compared to the natural substrate. This difference in efficiency was imparted by a 53-fold decrease in the k pol and a 4.5-fold decrease in the binding affinity. From an acyclic nucleotide 2, to the cyclobutyl ring 3, the efficiency was decreased by 4-fold, which was due to a 4-fold decrease in the k pol. I Details on the synthesis of these compounds have been reported separately. (Li Y, Hager MW, Mao S, Bluemling GR, Wang L, Kim J, Schinazi RF, Anderson KS, Liotta DC. Synthesis and anti-hiv activity of cyclobutyl nucleoside phosphonate analogs. ACS Med Chem Letters. 2012 Submitted) A brief synopsis of the synthesis is included in the Supporting Information.

Kim et al. Page 3 Many studies involving the presence of fluorine in nucleotide analogs have the substituent at the base of the nucleotide. Incorporation studies of these fluorine substituted analogs using a DNA/DNA primer-template have demonstrated an improvement in incorporation efficiency. 7 Lobucavir contains a hydroxy methyl substituent at the 2 position of the cyclobutyl ring. The fluorine present on at the 2' position of the cyclobutyl ring, 4, negatively affected the incorporation compared to its parent compound, 3, and was 2-fold less efficient (0.0052 s 1 µm 1 for 4) as a substrate for incorporation. This difference in efficiency was observed through a 2-fold decrease in k pol. Although the rate of incorporation was affected, the K d of 3 and 4 were within error. A phosphonyl-diphosphate form of the cyclobutyl adenosine analog was synthesized, 5. This compound was clearly a better substrate for incorporation by RT compared to the triphosphate form 3. The efficiency of incorporation for the phosphonyl diphosphate 5 was 20-fold higher (0.21 s 1 µm 1 ) compared to the triphosphate 3. This increase in efficiency was imparted by a 6-fold increase in the k pol as well as the 3-fold tighter binding affinity. A phosphonate group has several advantages over a phosphate. The phosphorous-carbon bond is not susceptible to hydrolysis by phosphatases. Also, once the prodrug form of a nucleoside has entered a cell, the the presence of the phosphonate group bypasses the first phosphorylation step, which is considered to be the rate limiting step in the conversion of the triphosphate form of a nucleoside by cellular kinases. A recent review discusses these advantages and gives examples of other antiviral phosphonate candidates. 8 Interestingly, compound 5 was 4.4-fold more efficiently incorporated by RT WT compared to the tenofovir diphosphate 2 (Figure 1, solid bars). Compound 5 had a 2-fold faster k pol and an almost 3- fold tighter binding affinity compared to the FDA approved nucleotide analog. Compound 5 was able to act as a chain terminator by inhibiting incorporation of the next correct nucleotide, as shown in Supp. Figure 1. Another cyclobutyl containing compound 6, was synthesized as an adenosine analog to base-pair with dttp. This compound was not a good substrate for incorporation by RT, and was almost 2-fold less efficient (0.007 s 1 µm 1 ) compared to the parent compound 3. This difference was due to the almost 2-fold decrease in the k pol. The HIV-1 RT s ability to discriminate a nucleotide analog was determined, which is the ratio of the efficiency of incorporation for the natural substrate 1 to the efficiency of incorporation for the analog. The RT WT was 4-fold less discriminative against compound 5 versus compound 2. The enzyme was 4 7 fold more discriminative against the other cyclobutyl analogs (3, 4, 6) versus compound 2, and 19 40 fold more discriminative versus compound 5. Tenofovir-diphosphate 2, is an acyclic adenosine analog that also contains a phosphonate group, similarly to compound 5. Upon administration of tenofovir to infected persons, a drug resistant form of HIV-1 RT may arise containing a K65R mutation. We determined the pre-steady state kinetic parameters for the natural substrate datp, 1, tenofovir diphosphate 2, and the most promising cyclobutyl compound, 5 using the K65R mutant form of HIV-1 RT, as shown in Table 2. The RT K65R was 4-fold less efficient in incorporating the natural substrate 1, and 14-fold less efficient in incorporating the cyclobutyl compound 5. The large decrease in incorporation efficiency using the mutant RT was observed through a decrease in the k pol, compared to the wild type enzyme. Compound 2 was 9-fold less efficiently incorporated by the mutant enzyme compared to the wild type which was imparted by a 5-fold decrease in the k pol, and a 2-fold decrease in the binding affinity. The RT K65R was able to incorporate the cyclobutyl compound 5 more efficiently compared to the tenofovir diphosphate 2

Kim et al. Page 4 (Figure 1, cross hatch bars) by 3-fold. The discrimination of the two nucleotide analogs was determined, which is the ratio of the efficiency of incorporation for the natural substrate 1 to the efficiency of incorporation for the analog. The RT K65R mutant was 3-fold less discriminative against compound 5 versus compound 2. Recent work has been performed to solve the crystal structures of wild type and the K65R mutant of RT in the presence of a primer-template and datp or TFV-DP. 9, 10 Three major residues are involved in the interactions with the incoming dntp in both polymerases K(R)65, R72, and Y115. R72 interacts with the base, sugar, and α-phosphate of the incoming nucleotide. When K65 is mutated to an R, guanidinium stacking occurs with this amino acid and R72, causing steric restriction on R72 and therefore affecting the incorporation rate by RT. The amino acid Y115 stacks against the ribose ring of the natural substrate, which is partially lost with TFV-DP due to its acyclic structure as shown in Figure 2a. In order to analyze potential binding interactions of 5, the compound was docked into the RT active site bound to primer-template (Tuske et al. reference 10, PDB code 1T05). The cyclobutyl ring of compound 5 may be able to stack with Y115, or form favorable Van der Waals contacts, which may explain its better binding affinity compared to TFV-DP as shown in Figure 2a and 2b. Similar to the natural substrate and TFV-DP, the K65R mutation greatly decreases the rate of incorporation perhaps in the same manner, through the restriction of the R72 and its interaction with the phosphonate group present in the cyclobutyl compound. It is interesting to note that from the wild type RT structure with TFV-DP, the side chains of Lys65 and Lys219 are positioned to make potential salt bridges with the α-phosphate of the phosphonate. Based on the binding predictions from our model, potential salt bridges may be formed between compound 5, Lys65, and Lys219 (Figure 2b). This differs from dntp in which only Lys65 is positioned to make a potential salt bridge with the α-phosphate. The additional salt bridge formed with the phosphonate may give rise to potency of the compound. The docking model and structural data may also help explain the increased catalytic efficiency of HIV-1 RT observed with the phosphonate compound 5 compared to the triphosphate compound 3. Several cyclobutyl adenosine triphosphate analogs were evaluated for their ability to be incorporated by HIV-1 RT using pre-steady state kinetics. These data indicate the potential for cyclobutyl compounds to be a substrate for the enzyme. The phosphonyl diphosphate cyclobutyl adenosine analog 5 was most efficiently incorporated with a similar K d to the natural substrate 1. Compound 5 was also more efficiently incorporated compared to the clinically available tenofovir diphosphate 2 with both the wild type and K65R mutant forms of HIV-1 RT. We also demonstrated the ability of compound 5 to act as chain terminator by inhibiting incorporation of the next incoming nucleotide. Future work will further explore the phosphonyl group and use tenofovir disoproxil fumarate and the compound GS7340, an isopropylalaninyl monoamidate phenyl monoester prodrug of TFV 11 as models for a prodrug strategy of compound 5. Supplementary Material Acknowledgments Refer to Web version on PubMed Central for supplementary material. We would like to thank Dr. Stephen Hughes, Dr. Paul Boyer and Dr. Andrea Ferris (NIH) for the HIV-1 RT WT clone. We would also like to acknowledge Sierra Bioresearch (Tucson, AZ) for synthesizing compound 4. We would also like to thank the National Institutes of Health for support to GM49551 to K.S.A. RFS is supported by CFAR NIH grant 2P30-AI-050409 and the Department of Veterans Affairs.

Kim et al. Page 5 References 1. Deval J, White KL, Miller MD, Parkin NT, Courcambeck J, Halfon P, Selmi B, Boretto J, Canard B. Mechanistic basis for reduced viral and enzymatic fitness of HIV-1 reverse transcriptase containing both K65R and M184V mutations. J Biol Chem. 2004; 279(1):509 516. [PubMed: 14551187] 2. White KL, Margot NA, Wrin T, Petropoulos CJ, Miller MD, Naeger LK. Molecular mechanisms of resistance to human immunodeficiency virus type 1 with reverse transcriptase mutations K65R and K65R+M184V and their effects on enzyme function and viral replication capacity. Antimicrob Agents Chemother. 2002; 46(11):3437 3446. [PubMed: 12384348] 3. Li Y, Mao S, Hager MW, Becnel KD, Schinazi RF, Liotta DC. Synthesis and evaluation of 2'- substituted cyclobutyl nucleosides and nucleotides as potential anti-hiv agents. Bioorg Med Chem Lett. 2007; 17(12):3398 3401. [PubMed: 17434736] 4. Kati WM, Johnson KA, Jerva LF, Anderson KS. Mechanism and fidelity of HIV reverse transcriptase. J Biol Chem. 1992; 267(36):25988 25997. [PubMed: 1281479] 5. Frangeul A, Barral K, Alvarez K, Canard B. In vitro suppression of K65R reverse transcriptasemediated tenofovir- and adefovir-5'-diphosphate resistance conferred by the boranophosphonate derivatives. Antimicrob Agents Chemother. 2007; 51(9):3162 3167. [PubMed: 17620380] 6. Suo Z, Johnson KA. Selective inhibition of HIV-1 reverse transcriptase by an antiviral inhibitor, (R)-9-(2-Phosphonylmethoxypropyl)adenine. J Biol Chem. 1998; 273(42):27250 27258. [PubMed: 9765248] 7. Ray AS, Schinazi RF, Murakami E, Basavapathruni A, Shi J, Zorca SM, Chu CK, Anderson KS. Probing the mechanistic consequences of 5-fluorine substitution on cytidine nucleotide analogue incorporation by HIV-1 reverse transcriptase. Antivir Chem Chemother. 2003; 14(3):115 125. [PubMed: 14521328] 8. De Clercq E. The clinical potential of the acyclic (and cyclic) nucleoside phosphonates: the magic of the phosphonate bond. Biochem Pharmacol. 2011; 82(2):99 109. [PubMed: 21501598] 9. Das K, Bandwar RP, White KL, Feng JY, Sarafianos SG, Tuske S, Tu X, Clark AD Jr, Boyer PL, Hou X, Gaffney BL, Jones RA, Miller MD, Hughes SH, Arnold E. Structural basis for the role of the K65R mutation in HIV-1 reverse transcriptase polymerization, excision antagonism, and tenofovir resistance. J Biol Chem. 2009; 284(50):35092 35100. [PubMed: 19812032] 10. Tuske S, Sarafianos SG, Clark AD Jr, Ding J, Naeger LK, White KL, Miller MD, Gibbs CS, Boyer PL, Clark P, Wang G, Gaffney BL, Jones RA, Jerina DM, Hughes SH, Arnold E. Structures of HIV-1 RT-DNA complexes before and after incorporation of the anti-aids drug tenofovir. Nat Struct Mol Biol. 2004; 11(5):469 474. [PubMed: 15107837] 11. Lee WA, He GX, Eisenberg E, Cihlar T, Swaminathan S, Mulato A, Cundy KC. Selective intracellular activation of a novel prodrug of the human immunodeficiency virus reverse transcriptase inhibitor tenofovir leads to preferential distribution and accumulation in lymphatic tissue. Antimicrob Agents Chemother. 2005; 49(5):1898 1906. [PubMed: 15855512]

Kim et al. Page 6 Figure 1. Incorporation Efficiency for deoxyadenosine triphosphate (datp), tenofovir diphosphate (TFV-DP), cyclobutyl adenosine phosphonyl disphosphate (5) for wild type (solid black) and K65R (cross hatch) HIV-1 RT. The natural substrate, datp, is the most efficiently incorporated. The cyclobutyl analog, 5, is more efficiently incorporated by both forms of RT compared to the FDA approved TFV-DP.

Kim et al. Page 7 Figure 2. Tenofovir and Compound 5 in the active site of HIV-1 RT. a) Crystal structure of HIV-1 RT cross-linked to template-primer and tenofovir-diphosphate bound as the incoming nucleotide substrate (PDB code: 1T05). b) Docked model of HIV-1 RT with compound 5 bound as the incoming nucleotide substrate using Autodock Vina. Information on the docking model is in the Supplementary Information.

Kim et al. Page 8 Table 1 Structures and pre-steady state kinetic data for incorporation of deoxyadenosine triphosphate, tenofovir diphosphate, and cyclobutyl analogs using HIV-1 RT WT Compound Structure k pol (s 1 ) K d (µm) Efficiency (s 1 µm 1 ) Discrimination 1 12.3 ± 1.0 4.6 ± 1.6 27 2 0.79 ± 0.08 16.5 ± 4.6 0.048 56 3 0.23 ± 0.02 20.4 ± 4.6 0.011 245 4 0.13 ± 0.01 25.5 ± 3.8 0.0052 519 5 1.3 ± 0.1 6.5 ± 2.0 0.21 13 6 0.16 ± 0.01 22.4 ± 2.8 0.007 386

Kim et al. Page 9 Table 2 Pre-steady state kinetic data for incorporation of deoxyadenosine triphosphate (datp), tenofovir diphosphate (TFV-DP) and cyclobutyl-adenosine-phosphonyl-diphosphate (5) using HIV-1 RT K65R Compound K pol (S 1 ) K d (µm) Efficienty (s 1 µm 1 ) datp 4.0 ± 0.3 6.6 ± 1.2 0.61 Discrimination TFV-DP 0.15 ± 0.01 29.1 ± 6.0 0.005 122 5 0.07 ± 0.004 4.7 ± 1.0 0.015 41