throbber
Q-D) 1994 Oxford University Press
`
`Nucleic Acids Research, 1994, Vol. 22, No. 20 4259-4267
`
`Termination of DNA synthesis by novel 3'-modified-
`deoxyribonucleoside 5'-triphosphates
`
`Michael L.Metzker*, Ramesh Raghavachari1 +, Stephen Richards, Swanee E.Jacutin1,
`Andrew Civitello, Kevin Burgess1 and Richard A.Gibbs
`Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza,
`Houston, TX 77030 and 1Department of Chemistry, Texas A & M University, College Station,
`TX 77843, USA
`
`Received June 3, 1994; Revised and Accepted July 25, 1994
`
`ABSTRACT
`Eight 3'-modified-dNTPs were synthesized and tested
`in two different DNA template assays for incorporation
`activity. From this enzymatic screen, two 3'-0-methyl-
`dNTPs were shown to terminate DNA syntheses
`mediated by a number of polymerases and may be used
`as alternative terminators in Sanger sequencing. 3'-0-
`(2-Nitrobenzyl)-dATP is a UV sensitive nucleotide and
`was shown to be incorporated by several thermostable
`DNA polymerases. Base specific termination and
`efficient photolytic removal of the 3'-protecting group
`was demonstrated. Following deprotection, DNA
`synthesis was reinitiated by the incorporation of natural
`nucleotides into DNA. The identification of this labile
`terminator and the demonstration of a one cycle stop-
`start DNA synthesis are initial steps in the development
`of a novel sequencing strategy.
`
`INTRODUCTION
`2'-Deoxyribonucleoside-5'-triphosphates (dNTPs) modified at
`their 3'-hydroxyl position can act as terminators of enzyme-
`directed DNA synthesis (1-12). These nucleotide analogs are
`probes,
`mechanistic
`as DNA sequencing tools,
`useful
`antimetabolites, and as antiviral agents. Consequently, such
`compounds have been used for analytical and therapeutic studies
`(2). Overall, however, the number of compounds that are well
`characterized is small, and there is considerable scope for new
`combinations of terminators and polymerases to be identified.
`Among the most familiar terminators of DNA synthesis are
`the 2', 3 '-dideoxyribonucleoside-5 '-triphosphates (ddNTPs) that
`are the basis for Sanger DNA sequencing (13). In that method
`oligonucleotide-primed DNA or RNA templates are enzymati-
`cally extended in a 5' - 3' direction in the presence of a mixture
`of dNTPs and ddNTPs to generate a population of molecules that
`are terminated at specific base positions. DNA fragments of
`different lengths are resolved by denaturing polyacrylamide gel
`
`electrophoresis and detected either by radioactive or fluorescent
`labels to reveal the underlying base sequence. Despite the obvious
`limitations of gel electrophoresis for sequencing long DNA
`strands, this method has been the favored approach for more than
`ten years (13,14).
`Improvements to the Sanger protocols are being sought to meet
`the increasing demands of large scale sequencing of whole
`genomes (14). We and others (15-18) have independently
`conceived a radically different, gel-free alternative to the Sanger
`scheme for DNA sequencing. This method, called the Base
`Addition Sequencing Scheme (BASS), is based on novel
`nucleotide analogs that terminate DNA synthesis. BASS involves
`repetitive cycles of incorporation of each successive nucleotide,
`in situ monitoring to identify the incorporated base, and
`deprotection to allow the next cycle of DNA synthesis, (Figure
`1). Compared to Sanger sequencing, BASS has two major
`advantages: base resolution would not require gel electrophoresis
`and there is a tremendous capacity for simultaneous analyses of
`multiple samples. The complete scheme demands nucleotide
`analogs that are tolerated by polymerases, spectroscopically
`distinct for each base, stable during the polymerization phase,
`and deprotected efficiently under mild conditions in aqueous
`solution. These stringent requirements are formidable obstacles
`for the design and synthesis of the requisite analogs.
`The investigation of the interactive patterns between various
`terminating analogs and different enzymes is an important
`preliminary phase in the development of the BASS method.
`Consequently, eight 3'-modified-dNTPs were synthesized and
`examined for their ability to terminate DNA synthesis mediated
`by a variety of polymerases. The majority of 3'-modified analogs
`have labile protecting groups that have the potential to be
`incorporated into BASS. Active combinations of terminators and
`enzymes were identified using two different primer-template gel
`assays. One of these compounds, 3'-0-(2-nitrobenzyl)-dATP [7],
`was used to demonstrate one complete cycle of termination,
`deprotection, and reinitiation of DNA synthesis.
`
`*To whom correspondence should be addressed
`+Present address: LI-COR Inc., Biotechnology Division, Lincoln, NE 68504-5000, USA
`
`Illumina Ex. 1039
`IPR Petition - USP 10,435,742
`
`

`

`4260 Nucleic Acids Research, 1994, Vol. 22, No. 20
`
`=UmHE
`
`Solid support
`
`'OPA
`
`3' OPC
`
`3' OPG
`
`3' OPT
`
`Base Addition
`and
`Termination
`
`/
`
`/ 3' OPA
`/lllllll
`A
`
`I
`
`k
`
`l~~~~~~IIIIIImIIII A/3OH
`
`Base
`Identification
`
`Base Protecting
`Group Removal
`
`Imaging of
`Reporter Group
`
`HO
`
`R
`O-
`
`9
`9
`O-V -O--Adenine
`O2- I
`0-
`
`where P' =
`
`Op
`
`/CH3
`
`1
`0
`
`>,jSCH3 /.CH2
`0
`2
`
`3
`
`H2N0
`
`H2N>:
`
`NO2
`5
`
`6
`9
`9
`R
`HO-{) PV-O-I
`0-
`0-
`
`OCH3
`8
`
`4
`
`7
`
`Figure 1. Cartoon of the Base Addition Sequencing Scheme (BASS). A primer
`is annealed to a biotinylated-labeled template bound to a solid support. Four
`deoxynucleotides triphosphates that have spectroscopically unique blocking groups
`attached to the 3'-position are added. Polymerase extension is terminated after
`the addition of one base. Upon imaging of the reporter group, the protecting group
`is removed resulting in a 'free' 3'-OH group, allowing the addition of the next
`complement base.
`
`MATERIALS AND METHODS
`General
`High field Nuclear Magnetic Resonance (NMR) spectra were
`recorded on a Bruker AC250 (1H at 250 MHz, 13C at 62.9
`MHz, 31P at 101.26 MHz) or a Varian XL 200 (1H at 200
`MHz, 13C at 50 MHz, 31P at 81 MHz). Ultraviolet (UV) spectra
`were recorded on a Hewlett-Parkard model 8452A diode array
`spectrophotometer. Thin layer chromatography was performed
`on Whatman silica gel 60 A F254 plates. Flash chromatography
`was performed on SP silica gel 60 (230-600 mesh ASTM). Ion-
`exchange chromatography was performed on Fluka DEAE
`cellulose C451 (HCO3- form). Photodecomposition of 3'-O-
`(2-nitrobenzyl)-dATP [7] was performed using a FisherBiotech
`transilluminator.
`
`Organic syntheses
`The chemical structures of compounds [1] -[8] are shown in
`Figure 2. Compounds [1]- [4], [6], [8] were prepared according
`to the general scheme:
`
`HO
`
`Base
`
`t-BuPh2SiC1
`
`TBDPSO
`
`Base
`
`OH
`
`imidazole, DMF
`
`add 3-group
`
`TBDPSO
`
`Base
`
`Bu4NF
`
`OH
`
`9
`
`HO s
`
`Bjlase
`
`OH
`OH
`The 5'-hydroxyl was protected with a tert-butyldiphenylsilyl
`(TBDPS) group, and the specific addition of the 3'-protecting
`
`THF
`
`Figure 2. Chemical structures of the 3'-modified-nucleotides. Details of the
`chemical syntheses are described in the Materials and Methods section.
`
`groups (P') are described below. Desilylations were performed
`by the addition of 1.0 equiv. of tetrabutylammonium fluoride
`(Bu4NF) to the 3'-protected-5'-silyl-adenosine or thymidine
`derivatives. The reactions were monitored by TLC; after
`completion (ca. 15 min.), the reactions were quenched with 1.0
`equiv. of glacial acetic acid. The solvent was removed, and the
`residues were purified by silica column chromatography (10%
`methanol/ethyl acetate).
`
`[1]. To 2'-deoxy-5 '-tert-
`2 '-Deoxy-3 '-0-methyladenosine
`butyldiphenylsilyladenosine [9] (200 mg, 0.4 mmol) in benzene
`(5 mL), methyl iodide (568 mg, 4.0 mmol, 10 equiv.),
`tetrabutylammonium hydroxide (TBAH) (40% solution, 325 ,uL),
`and 1 M NaOH (5 mL) were vigorously stirred at 25°C for 16
`h. The organic layer was extracted with ethyl acetate and washed
`with deionized (D.I.) water, saturated NaCl, dried over Na2SO4
`and purified by flash chromatography using a stepwise gradient
`(0% methanol/ethyl acetate to 5% methanol/ethyl acetate in 2%
`intervals) (180 mg, 89%) (19).
`The 0-methyl derivative from the above procedure (80 mg,
`0.16 mmol), after desilylation and flash chromatography gave
`compound [1] as colorless crystals (30 mg, 70%). High resolution
`mass spectrometry (HRMS) m/e calculated for C11H15N503:
`265.1172, observed 265.1154.
`
`2'-Deoxy-3'-O-acyladenosine
`[2].
`2'-Deoxy-5'-tert-butyldi-
`phenylsilyladenosine [9] (100 mg, 0.2 mmol), acetic anhydride
`(28 mg, 0.27 mmol), and 4-dimethylaminopyridine (DMAP) (5
`mg, 0.05 mmol) in dry pyridine were stirred at 25°C for 6 h.
`After removing pyridine under vacuum, the residue was dissolved
`in D.I. water, extracted in chloroform, washed with D.I. water,
`10% HCI, saturated NaHCO3, saturated NaCl, dried over
`Na2SO4 and flash chromatographed (96 mg, 90%).
`The 3'-O-acyl derivative (100 mg, 0.19 mmol) following
`desilylation and flash chromatography afforded compoud [2] (44
`
`

`

`mg, 80%). HRMS mie calculated for C12H15N504: 293.1121,
`observed 293.1107.
`
`2'-Deoxy-5 '-tert-butyldi-
`[3].
`2 '-Deoxy-3 '-0-allyladenosine
`phenylsilyladenosine [9] (200 mg, 0.4 mmol) in benzene (5 mL),
`allyl bromide (484 mg, 4.0 mmol, 10 equiv.), TBAH (40%
`solution, 390 MLL), and 1 M NaOH (5 mL) were stirred at 25°C
`for 15 h. Following ethyl acetate extraction, the organic phase
`was washed with D.I. water, saturated NaCl, dried over
`Na2SO4, and flash chromatographed (157 mg, 74%).
`The O-allyl derivative (198 mg, 0.37 mmol) following
`desilylation and flash chromatography gave compound [3] (106
`mg, 98.5%). HRMS mie calculated for C13H17N503: 291.1328,
`observed 291.1318.
`
`2 '-Deoxy-3 '-O-tetrahydropyranyladenosine [4]. 2'-Deoxy-5'-tert-
`butyldiphenylsilyladenosine [9] (2.90 g, 5.92 mmol), dihydro-
`pyran (4.89 g, 59.2 mmol, 10 equiv.) and pyridinium nitro-
`benzenesulfonate (1.67 g, 5.92 mmol) were dissolved in
`methylene chloride (20 mL) and stirred at 40°C for 20 h. The
`reaction mixture was washed with D.I. water, saturated NaCl,
`dried over Na2SO4 and flash chromatographed to give a
`diastereomeric mixture of 2'-deoxy-3'-O-tetrahydropyranyl-5'-
`tert-butyldiphenylsilyladenosine (0.4 g, 12%).
`The tetrahydropyran derivative formed above, after desilylation
`and flash chromatography yielded compound [4] (147 mg, 84%)
`as a mixture of diastereomers. HRMS m/e calculated for
`C15H21N504: 335.1589, observed 335.1581.
`
`2 '-Deoxy-3 '-O-(2-aminobenzoyl)adenosine-5 '-triphosphate [5].
`This compound was prepared according to the procedure of
`Hiratsuka et al. (20) directly from the 2'-deoxyadenosine-5'-tri-
`phosphate sodium salt.
`2'-Deoxy-3'-O-(4-nitrobenzoyl)adenosine [6]. 2'-Deoxy-5'-tert-
`butyldiphenylsilyladenosine [9] (100 mg, 0.2 mmol), 4-nitro-
`benzoyl chloride (89 mg, 0.48 mmol), DMAP (5 mg, 0.04 mmol)
`were dissolved in pyridine and stirred for 8 h. Following solvent
`removal, the residue was dissolved in chloroform and was washed
`D.I. water, saturated NaHCO3, D.I. water, saturated NaCl,
`dried over Na2SO4, and flash chromatographed giving a
`colorless solid (73 mg, 57%).
`The 4-nitrobenzoyl derivative (66 mg, 0.1 mmol) following
`desilylation and flash chromatography gave compound [6] (22
`mg, 57%). HRMS mie calculated for C17H16N606: 400.1128,
`observed 400.1140.
`
`2'-Deoxy-3'-0-(2-nitrobenzyl)adenosine [7]. 2'-Deoxyadenosine
`(100 mg, 0.4 mmol) [dried by repeated coevaporation with
`pyridine] was dissolved in hot DMF and cooled to 0°C in an
`ice bath. To the above solution, NaH (26 mg, 0.52 mmol [50%
`in mineral oil] in DMF after washing with dry benzene was added
`and stirred for 45 min. 2-Nitrobenzyl bromide (95 mg, 0.44
`mmol) in DMF was added, and the reaction stirred for 3 h. The
`reaction was quenched with cold D.I. water and stirred overnight.
`The solid obtained was filtered, dried, and recrystalized in ethanol
`(122 mg, 79%). HRMS mie calculated for C17H18N605:
`386.1335, No mie was observed. Fast atom bombardment MS,
`nitrobenzyl alcohol (NBA) mle 387.1 (M +1).
`2'-Deoxy-3'-O-methylthymidine [8]. 2'-Deoxy-5'-tert-butyldi-
`phenylsilylthymidine [91 (100 mg, 0.21 mmol) in benzene (5 mL),
`
`Nucleic Acids Research, 1994, Vol. 22, No. 20 4261
`
`methyl iodide (43 mg, 0.3 mmol), TBAH (40% solution, 325
`ML), and 1 M NaOH (5 mL) were vigorously stirred at 25°C
`for 6 h. The organic layer was extracted with ethyl acetate and
`washed with D.I. water, saturated NaCl, and dried over Na2SO4
`(100 mg, 98%).
`The above sample, following desilylation and purification by
`flash chromatography, gave compound [8] (36 mg, 88%). HRMS
`mie calculated for C1IH16N205: 256.1059, observed 256.1082.
`Syntheses of nucleoside 5'-triphosphates
`In general, the 3'-modified nucleoside (1.0 equiv.) was dissolved
`in trimethylphosphate under nitrogen atmosphere. Phosphorus
`oxychloride (POCl3) (3.0 equiv.) was added, and the reaction
`stirred at - 10°C for 4 h. The reaction was quenched with a
`solution of tributylammonium pyrophosphate (5.0 equiv.) in
`DMF and tributylamine (0.2 mL) (21). After stirring vigorously
`for 10 min., the reaction was quenched with 2 mL of 2 M TEAB,
`pH 7.5. The solution was concentrated, and the triphosphate
`derivative was isolated by linear gradient (0.01 M to 0.5 M
`TEAB) using a DEAE cellulose (HCO3- form) column.
`Reverse-phase high performance liquid chromatography
`(RP-HPLC)
`The RP-HPLC hardware system consisted of a Beckman
`controller and model 100A pumps, a Rheodyne model 7125
`injector, an Applied Biosystems (ABI) model 759A absorbance
`detector, and a Spectra-Physics model SP4600 DataJet integrator.
`Gradient RP-HPLC was performed using an ABI aquapore
`OD-300 column (4.6 mmx250 mm) where 'Buffer A' is 100
`Buffer
`mM triethylammonium acetate (TEAA), pH 7.0 and
`B' is 100 mM TEAA, 70 % (v/v) acetonitrile. Compounds
`[1]-[6] and [8] were purified using the following gradient
`conditions: 0% B, 5 min.; 0% B - 40% B, 60 min.; 40% B
`- 100% B, 18 min.; 100% B, 5 min. at a flow rate of 0.5 mL
`per min. Compound [7] was purified using the following gradient
`conditions: 30% B, 5 min.; 30% B - 70% B, 60 min.; 70%
`B -100% B, 15 min.; 100% B, 5 min. The gradient conditions
`used to analyze individual nucleotides were: 0% B, 5 min.; 0%
`B - 40% B, 30 min.; 40% B - 100% B, 18 min.; 100% B,
`5 min.
`
`Polymerases
`Avian Myeloblastosis Virus (AMV) and Moloney Murine
`Leukemia Virus (M-MuLV) reverse transcriptases, Klenow
`fragment of DNA polymerase I, and T4 polynucleotide kinase
`were purchased from Pharmacia. Bst DNA polymerase was
`purchased from Bio-Rad Laboratories. AmpliTaqe DNA
`polymerase and rTth DNA polymerase were purchased from
`Perkin Elmer. Sequenase® was purchased from United States
`Biochemical. VentR®
`(exo-) DNA polymerase was kindly
`provided by New England Biolabs. Pfu (exo-) DNA
`polymerase was purchased from Stratagene.
`DNA templates
`M13mpl9 DNA was obtained from a 250 mL culture by
`polyethylene glycol precipitation and purified by a QIAGEN-tip
`100 column according to the manufacture's protocol. Universal
`primer (5'-TGTAAAACGACGGCCAGT), biotinylated and
`unbiotinylated oligonucleotide template (5 '-TACGGAGGTGG-
`ACTGGCCGTCGTTTTACA) and biotinylated oligonucleotide
`template (5'-TACGGAGGTTTTTGGACTGGCCGTCGTTT-
`
`

`

`4262 Nucleic Acids Research, 1994, Vol. 22, No. 20
`
`ACA) were synthesized using an ABI model 380B DNA
`synthesizer and purified by trityl-on RP-HPLC. All
`nonradioactive nucleotides were purchased from Pharmacia, and
`['y-32P]ATP was purchased from Amersham.
`Polymerase incorporation assays
`Two different template assays were used to test for 3 '-modified
`nucleotide incorporation. In the first, designated the 'M13mpl9-
`template assay', [32P]-labeled universal primer was annealed to
`single-stranded M13mpl9 DNA (0.1 pmol to 0.45 ,ug respect-
`ively, per 5 AL) in the specific enzyme buffer by heating to 80°C
`for 5 min. and cooling slowly to 25°C. The subsequent enzymatic
`extension of the primer-template complex was performed under
`conditions that are analogous to Sanger sequencing, where the
`natural nucleotides were mixed with either a dideoxynucleotide
`or 3'-modified nucleotide terminator to generate a sequencing
`ladder. For the second assay, designated the 'Oligo-template
`[32P]-labeled universal primer was annealed to an
`assay',
`oligonucleotide template (0.05 pmol to 0.1 pmol respectively,
`per 5 AL) in the same fashion. Subsequent extensions were
`performed in the absence of the natural nucleotide when either
`a dideoxynucleotide or 3'-modified nucleotide was tested.
`For each reaction, 5 ,uL aliquots of the annealed primer-
`template samples were dispensed into separate tubes containing
`5 liL mixtures of each enzyme and nucleotides in their specific
`buffers. The final buffer conditions, concentrations of nucleotides,
`enzymatic units, and incubation temperatures are given in Table
`1. The reactions were incubated for 10 min. and then stopped
`by the addition of 5 /sL of stop solution containing 98% D.I.
`
`formamide, 10 mM EDTA, pH 8.0, 0.025% bromophenol blue,
`and 0.025% xylene cyanol. The samples were heated to 85°C
`for 3 min., chilled on ice, and either 4 ,uL (M13mpl9-template
`Assay) or 3 izL (Oligo-template assay) were loaded on a 10%
`or 20% polyacrylamide gel,
`respectively. Following
`electrophoresis, the gel was fixed in an aqueous 10% acetic acid,
`10 % methanolic solution (v/v), dried, and autoradiographed on
`HyperfilmTM-MP (Amersham).
`Biotinylated Oligo-template assay
`The conditions of this assay are similar to the Oligo-template
`assay except prior to primer annealing, 2.0 pmol of biotinylated
`template was captured on 10 mL streptavidin coated magnetic
`M-280) in 1 M NaCl for 15 min.
`beads (Dynal Dynabeads®
`After washing the bound template in the specific enzyme buffer,
`0.1 pmol of [32P]-labeled universal primer was annealed to an
`oligonucleotide template and extension reactions were performed
`as described in the Oligo-template assay.
`
`RESULTS
`Syntheses and purification
`Compounds [1] through [8] were synthesized, desilylated,
`phosphorylated and purified as described above. In general, the
`yields of 3'-protection reactions ranged from 57% to 98% with
`the exception of 3'-O-THP-dATP [4]. This low yield (12%) is
`believed to be due to the acidic properties of the silica gel
`hydrolyzing the linkage to the THP group. For the desilylation
`reactions, the yields ranged from 70% to 98%, except for 3'-O-
`
`Table 1. Specific enzymatic conditions for both the M13mpl9-template and Oligo-template assays
`Bst DNA AmpliTaqe
`Klenow
`AMV-RT
`M-MuLV-RT
`Enzymatic
`SequenaseQD
`polymerase
`fragment
`DNA
`Conditions
`polymerase
`
`Pfu(exo-)
`DNA
`polymerase
`
`rTth DNA
`polymerase
`
`Vent(exo-)G
`DNA
`polymerase
`
`Buffers
`[Tris-HCl]
`[MgC12I
`
`50 mML pH 8.3 50 nmML pH 8.3
`8 mm
`8 mM
`10 mM DTT
`30 mM KCI
`
`10mM, pH 8.5 40nmL pH 7.5 10 mh,pH 8.5 10nML pH 8.5 20 niML pH 8.75
`5mM
`5mM
`10mM
`10omM
`2 mM MgSO4
`50 mM NaC
`50 mM NaCI
`10 mM KCI
`10 mM (NH)2SO4
`0.1% Triton X-100
`0.1 mg/mL BSA
`
`50 mhL pH 8.3 20mM, pH&8
`8mM
`2 ml MgSO4
`10 mM KG
`10rnMDTT
`10 mM (NH4)2SO4
`0.1% Triton X-100
`
`Incubation
`Temperature ('C)
`
`Units
`
`M13mpl9-Templ.
`[dATPi (pM)
`[dCTPI (uM)
`[dGTPl (,M)
`IdTTPl (o)
`[ddATPI (W)
`[ddCTPI (riM)
`[ddGTPI (pM)
`[ddTTPI (&tM
`
`37
`
`1.3
`
`40
`40
`40
`40
`2
`2
`2
`2
`
`37
`
`1.9
`
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`
`37
`
`1.0
`
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`
`37
`
`1.3
`
`10
`10
`10
`20
`0.25
`0.25
`0.25
`0.5
`
`65
`
`0.1
`
`1
`1
`1
`1
`10
`10
`10
`10
`
`68
`
`0.3
`
`2.5
`2.5
`2.5
`2.5
`150
`75
`15
`150
`
`Oligo-T*mpl.
`[dATPJ (MM)
`0.5
`2.5
`0.5
`2.5
`0.025
`0.25
`[dCTPJ (pM)
`0.5
`2.5
`2.5
`0.5
`0.025
`0.25
`[dTTPJ (ps)
`2.5
`2.5
`0.5
`2.5
`0.025
`0.25
`[ddATP] (M)
`250
`2.5
`0.05
`2.5
`0.25
`2.5
`[ddGTPJ (g)
`0.5
`250
`0.05
`2.5
`0.25
`0.25
`[ddTTPl (,)
`5
`5
`0.5
`2.5
`25
`2.5
`N/D means the assay conditions were not determined and I/T means ddNTP termination was incomplete.
`
`75
`
`0.1
`
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`
`2.5
`2.5
`2.5
`I/T
`I/T
`I/T
`
`74
`
`0.3
`
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`
`0.5
`2.5
`0.5
`125
`25
`100
`
`72
`
`0.1
`
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`N/D
`
`0.5
`0.5
`0.5
`125
`25
`100
`
`

`

`(4-nitrobenzoyl)-dATP [6]. The yield was reduced in that case
`to 57% due probably to a rearrangement of the 4-nitrobenzoyl
`group from the 3'-position to the 5'-position (data not shown).
`The phosphorylation yields of compounds [1]- [4] and [6]- [8]
`ranged from 25% to 40%. Thymidine analogs including 3'-O-
`methyl-dTTP [8], however, had to be handled more cautiously
`since they were more rapidly degraded by tributylammonium
`pyrophosphate than were the adenosine analogs.
`The 3'-modified-dNTPs were further purified by RP-HPLC
`to 2 99% prior to the polymerase assay. Each nucleotide
`synthesis initially contained several major peaks that were
`individually tested in the Oligo-template assay to determine the
`active species. In general, the adenosine analogs contained both
`the natural dATP and 3'-modified-dATP.
`
`Termination assays
`A series of polymerases were chosen to test the candidate
`based on their broad template
`terminators,
`3 '-modified
`specificities and their commercial availability. The conditions for
`screening compounds [1]- [8] using each enzyme were first
`defined by a series of control polymerization experiments. For
`the M13mpl9-template assay, a range of dNTP and ddNTP
`concentrations was identified that gave a clear sequencing ladder.
`Each test gel subsequently contained constant dNTP/ddNTP ratios
`in control lanes for three bases, while the concentrations of the
`test compound and its corresponding ddNTP were varied.
`The Oligo-template assay was also standardized before testing
`each 3'-modified-dNTP for termination. The synthetic template
`contained all four bases to allow for the incorporation of the
`remaining natural nucleotides, so that other aspects of the enzyme
`performance could be identified. We found that
`all the
`polymerases misincorporated other dNTPs in the absence of the
`complement dNTP, and this nucleotide readthrough was
`concentration dependent. Thus, minimum dNTP concentrations
`that gave efficient incorporation, but no apparent misincorporation
`were first defined in this assay. These dNTP concentrations were
`then used to determine the minimum ddNTP concentration that
`yielded complete termination.
`(exo-) DNA polymerase was
`excluded from the Oligo-template assay since a ddNTP
`concentration that yielded complete termination for this enzyme
`
`Nucleic Acids Research, 1994, Vol. 22, No. 20 4263
`
`could not be identified. In each of the Oligo-template gels, the
`reactions contained all the required nucleotides except the natural
`nucleotide corresponding to the analog tested. The samples
`routinely used in this assay were a blank control (absence of
`corresponding dNTP or ddNTP), a titration of the corresponding
`ddNTP, a readthrough control (presence of corresponding
`dNTP), and a titration of the corresponding 3'-modified-dNTP.
`
`Terminator screen
`Table 2 summarizes the data from the enzymatic screen of
`compounds [1] - [8]. Three main classes of activity were defined:
`termination, inhibition, and inactive. Termination was apparent
`when the reaction containing the test compound mimicked the
`migration pattern of the ddNTP control. Inhibition was revealed
`when the presence of the test compound prevented the polymerase
`from incorporating the natural nucleotides. No activity was
`recorded when the 3'-modified-dNTPs mimicked either the blank
`or readthrough controls. In addition, a fourth effect related to
`an alteration in enzymatic fidelity is discussed below. Compounds
`[1], [8], and [7] showed specific termination and were further
`evaluated with respect to their concentration dependent effects.
`3'-O-methyl-dATP [1] incorporation
`in Figure 3A shows the
`The M13mpl9-template assay
`incorporation of 3'-O-methyl-dATP [1] by AMV-RT. The
`termination of DNA synthesis by 3'-O-methyl-dATP [1] mimics
`the ddATP controls in a concentration dependent manner,
`although each band appears to migrate slightly slower. From the
`comparison of termination band intensities, it can be estimated
`that 3'-O-methyl-dATP [1] is approximately 200 to 250-fold less
`efficiently incorporated by AMV-RT than ddATP (compare lanes
`6 and 8: 5 mM and 1 mM, respectively). In Figure 3B, the Oligo-
`template gel also shows the incorporation of 3 '-O-methyl-dATP
`[1] by AMV-RT. In addition to termination, some readthrough
`was also observed due to the presence of contaminating dATP.
`All RP-HPLC purified 3'-modified-dATPs (compounds [1]- [7])
`showed approximately 1% dATP contamination, and these trace
`levels could not be removed by subsequent RP-HPLC.
`3'-O-Methyl-dATP [1] was also incorporated by M-MuLV-
`RT and inhibited DNA syntheses by rTth and VentR® (exo-)
`
`Table 2. Activity matrix of RP-HPLC purified 3'-protecting dNTPs challenged against commercially available polymerases
`AmpliTaq@ VentR(exo-)P
`Kienow
`3'-modified-dATP
`%qumase9
`rTth DNA
`Bst DNA
`AMV-RT
`M-MuLV-RT
`(except compound [81)
`polymerase
`polymerase
`fragment
`DNA
`DNA
`polymerase
`polymerase
`
`[11 0-methyl
`[21 0-acyl
`
`[3] 0-allyl
`
`[4] 0-tetrahydropyran
`[5l 0-(4-nitrobenzoyl)
`[6] 0-(2-anmnobenzoyl)
`
`[7] 0-(2-nitrobenzyl)
`[8] 3'-0-methyl-dTTP
`
`Termination Termination*|
`
`|
`
`|
`
`Ihibition
`
`Inhibition*
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`hihibition
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`Inhibition
`
`Termination*
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`-
`
`hIhibition
`
`Termination Termination* Termination
`
`-
`
`Inhibition
`
`Temiination Termination Termination Termination
`
`All compounds were assayed at a final concentration of 250 AM according to the conditions specified in Table 1. '-' means
`no activity was detected, 'Termination' means that the termination bands mimic ddNTP termination bands, and 'Inhibition'
`means the rate of DNA synthesis is reduced in a nonspecific manner. '*' means the activity was incomplete at a final concentration
`of 250 AM .
`
`

`

`j.........._
`I,b,,l,.
`!_.._....
`,,,., ;;t _..{_
`!bss*t_
`
`.h.x
`
`___
`
`......S
`
`4264 Nucleic Acids Research, 1994, Vol. 22, No. 20
`
`*ww s
`
`s_ w
`
`_ .7s,
`
`..\
`
`B.l
`11tkro,.~i
`:.coct
`
`rr. TI,
`
`rir-_
`
`a,
`
`':
`
`obtained by using the AMV-RT M13mpl9-template assay (data
`not shown). However, 3'-O-methyl-dTTP [8] was efficiently
`incorporated by AmpliTaq®
`DNA polymerase in the
`M13mpl9-template assay, and the termination pattern mimicked
`the ddTTP DNA ladders in a concentration dependent manner,
`(Figure 4A). Additional bands, however, were observed in the
`3'-O-methyl-dTTP [8] ladders that were generated by both
`AmpliTaqe DNA polymerase (see arrows) and Bst DNA
`polymerase (data not shown) assays. The position of these
`additional bands corresponded to ddATP termination bands
`suggesting misincorporation of the thymidine analog in place of
`the deoxyadenosine analog. The efficiency of incorporation
`relative to ddTTP was, therefore, not determined in the
`Ml3mp 19-template assays because of the additional bands.
`Figure 4B shows the results of challenging 3'-O-methyl-dTTP
`[8] against four thermostable DNA polymerases in the Oligo-
`template assay. 3'-O-Methyl-dTTP [8] terminates all four
`polymerases in a concentration dependent manner. From the
`comparison of band intensities of 3'-0-methyl-dTTP [8] to ddTTP
`termination products, it can be estimated that 3'-O-methyl-dTTP
`[8] is approximately 50-fold less efficiently incorporated by Bst
`efficiently
`DNA polymerase than ddTTP, 20-fold
`less
`incorporated by AmpliTaqe DNA polymerase than ddTTP,
`2-fold less efficiently incorporated by rTth DNA polymerase than
`ddTTP, and 10-fold less efficiently incorporated by VentR®
`(exo-) DNA polymerase than ddTTP.
`Further investigation of a different nucleotide composition in
`the Oligo-template assay revealed altered base specificity of 3'-0-
`methyl-dTTP [8]. In the absence of dATP, 3'-O-met1Vl-dTTP
`[8] was incorporated by both Bst and AmpliTaq
`DNA
`polymerases, and the termination bands mimicked the ddATP
`controls, (Figure 4C). It is noteworthy that in addition to 3'-O-
`methyl-dTTP [8] incorporation in Figure 4C, significant levels
`of readthrough were observed. Since the Oligo-template assay
`was performed in the absence of a deoxyadenosine analog, the
`readthrough must reflect the misincorporation of dNTPs in the
`noncomplement base position. This result suggests that 3'-O-
`methyl-dTTP [8] alters the base specific properties of DNA
`polymerases, not only in its incorporation, but in the incorporation
`of other natural nucleotides present in the reaction. This result
`also highlights the importance of the precise conditions of
`incorporation assays.
`
`3'-0-(2-nitrobenzyl)-dATP [7] incorporation by Bst DNA
`polymerase
`3'-O-(2-Nitrobenzyl)-dATP [7] terminated Bst DNA synthesis
`in a base specific manner. In the Bst M13mpl9-template assay,
`however, the termination did not correspond to any of the ddNTP
`controls, (Figure SA). This result made it difficult to assign a
`specific mode of action to 3'-O-(2-nitrobenzyl)-dATP [7]. The
`assignment was resolved using the biotinylated Oligo-template
`assay that gave strong evidence for termination. As shown in
`Figure 5B, 3'-O-(2-nitrobenzyl)-dATP [7] terminates Bst DNA
`synthesis where the DNA fragment migrates slower than the
`ddATP control, (compare lanes 3 and 7). 3'-O-(2-Nitrobenzyl)-
`dATP [7] is a UV sensitive compound that undergoes an
`intramolecular rearrangement by irradiation to give dATP and
`nitrosobenzaldehyde (22). Following UV exposure, the DNA
`termination fragment migrated at the same rate as the ddATP
`control, (compare lanes 3 and 8). This observation suggests that
`the 2-nitrobenzyl group can significantly alter the mobility of
`DNA in the gel and may provide a possible reason for the
`
`_.. ,,,,_.......
`
`_ wa:e.a_
`
`.|_
`_v
`
`_t
`
`tS
`
`.... _ ._.
`
`..
`
`, _
`
`_ _.
`
`....
`
`_M1_
`,
`
`0,_0...,*__ __
`r _s3?#s:
`::::_
`_ _
`
`__
`3x _s
`
`s_
`
`w
`
`- _
`
`,_
`
`_A
`
`_W
`
`_
`
`_
`
`_.A_f
`
`w
`
`_r
`
`C
`*
`_
`
`.
`
`.__ ___
`
`_
`
`-
`
`_ _P vw
`
`Incorporation of 3'-O-methyl-dATP by AMV-RT. (A)
`Figure 3.
`Ml3mpl9-template assay: In addition to the conditions specified in Table 1, lane
`1 contained no ddNTPs, and lanes 2-5 contained ddTTP, ddGTP, ddCTP, and
`ddATP, respectively. Lanes 6 and 7 contained 5 /AM and 10 zM of ddATP,
`respectively. Lanes 8-10 contained 1 mM, 5 mM, or 10 mM of 3'-O-methyl-
`dATP, respectively. (B) Conditions for the AMV-RT Oligo-template assay were
`used where lanes 1 and 6 contained no dNTPs or ddNTPs. Lanes 2-5 contained
`dATP, dCTP and ddGTP. In addition, lane 3 contained ddTTP, lane 4 contained
`dTTP, and lane 5 contained 500 itM of 3'-O-methyl-dTTP. Lanes 7-14 contained
`dCTP, dTTP and ddGTP. In addition, lane 8-10 contained 0.1 M, 0.5 ZM,
`and 2.5 ,sM of ddATP; lane 11 contained dATP; and lanes 12-14 contained
`10 ItM, 50 IM, and 250 jAM of 3'-O-methyl-dATP.
`
`DNA polymerases. In the M-MuLV-RT Oligo-template assay,
`we observed a batch-to-batch difference in assaying both 3'-O-
`methyl-dATP [1] and 3'-O-acyl-dATP [2], (data not shown).
`While, the ddNTP controls gave similar results in both M-MuLV-
`RT batch assays, 3'-O-methyl-dATP [1] showed minor
`termination, and both 3'-O-dATP analogs ([1] and [2]) showed
`partial inhibition of DNA synthesis in M-MuLV-RT batch 1. In
`contrast, 3'-O-methyl-dATP [1] showed significant termination
`of DNA synthesis, and both 3'-O-dATP analogs ([1] and [2])
`showed significant readthrough in M-MuLV-RT batch 2. This
`observation illustrates the importance of assaying candidate
`compounds with multiple enzyme batches.
`3'-O-methyl-dTTP [8] incorporation
`Unlike 3'-O-methyl-dATP [1], 3'-0-methyl-dTTP [8] was not
`incorporated by AMV-RT, (Figure 3B). A similar result was
`
`

`

`A.
`
`B.
`
`Bst DNA polymerase
`I4-.--.
`*
`
`AmpliTaqg DNA polymerase
`1 -
`4....
`r.
`r.
`
`234567 9a
`4
`
`..
`
`.123568
`
`4miwoo.
`
`Termination
`
`0
`
`Z
`
`f'W _
`
`1
`
`2 3 4 5 6
`
`7 8 9
`
`ddll P
`
`3 -0-methyl-dITI
`
`1 2 34 5 6 7 89
`ddTTP
`3 -D-methyl-dTl'
`
`I
`
`VentRr Cexoo) DNA polymerase
`
`rT1h DNA polymerase
`
`a-a_
`
`150
`
`4.s
`
`4.
`
`_
`
`1 2 3 4 5 6 7 8 9
`lT
`l'
`ddTTP
`3-O-methvy-dTTP
`
`1 2 3 4 5 6 7 8 9
`L llt
`l-
`ddTTP
`3'-O-methyl-dTTP
`
`C.
`
`Bst DNA polymnerase
`
`AmpliTaqg DNA polymeras
`
`...Readthrouph
`
`Ternination
`
`b
`
`.4.*
`
`-
`
`_
`681
`
`-3
`
`50
`
`1 2 3 4567 89
`ddA-T-P LT
`3'-D-methyl-dTTP
`
`12 34 56 7 89
`
`ddATP
`3'-O-methyl-dT1P
`
`Figure 4. Incorporation of 3'-O-methyl-dTTP by Bst, AmpliTaq®, rTth, and
`(exo-) DNA polymerases. (A) M13mpl9-template assay (AmpliTaq® ):
`VentR®
`Enzymatic conditions in Table 1 were used where lane 1 contained no ddNTPs,
`and lanes 2 -5 contained ddATP, ddCTP, ddGTP, and ddTTP, respectively. Lanes
`6 and 7 contained 450 ,AM and 750 of /oM ddTTP, respectively. Lanes 8-10
`contained 1 mM, 2.5 mM, or 5 mM of 3'-O-methyl-dTTP, respectively. Arrows
`correspond to termination bands that are observed in the ddATP control (compare
`lanes 2 with 8-10). (B) Conditions for the Oligo-template assay were used for
`Bst, AmpliTaq®, rTth , and VentR®
`(exo-) DNA polymerases. Lane 1
`contained no dNTPs or ddNTPs. Lanes 2-7 contained dATP, dCTP and ddGTP.
`In addition, lanes 3-5 contained (Bst ) 0.1 MM, 0.5 sAM and 2.5 IM ddTTP;
`(AmpliTaq® ) 1.0 MM, 5.0 AM and 25 AM ddTTP; (rth ) and (VentR®
`(exo-))
`4 IAM, 20 MM, and 100 AM ddTTP, respectively; lane 6 contained dTTP; and
`lanes 7-9 contained (Bst ) 4 MM, 20 MM, and 100 AM of 3'-O-methyl-dTTP;
`(exo-)) 20 AM, 100 AM, and 500 AM of
`(Ampl

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket