A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

Embed Size (px)

Citation preview

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    1/140

    UNIVERSITY OF SOUTHAMPTON

    FACULTY OF NATURAL AND ENVIRONMENTAL SCIENCES

    School of Chemistry

    MChem 3rd Year Dissertation

    A STRATEGY FOR ADDING MULTIPLE REPORTER GROUPS TO

    OLIGONUCLEOTIDES

    by Thomas Fleming

    Supervisors:

    Dr. Nittaya GaleProfessor Tom Brown

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    2/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    3/140

    Abstract

    Advances in DNA functionalisation impact on medical, material and

    forensic science, bio, nano and gene technology.1 Oligonucleotides as genetic

    probes offer rapid detection of complementary sequences with sufficient

    sensitivity to distinguish between sequences containing single base pair

    mismatches and normal gene sequences. Fluorescent labelling is a central

    feature of probe technologies. Single labelling is easily accomplished, but with

    increasingly sophisticated probes and demanding application conditions, such as

    sub-femtomolar target sequence concentrations, the demand for a convenient,

    effective and flexible, multiple internal labelling strategy is of rising importance.

    Current labelling strategies commonly use expensive dye-functionalisedphosphoramidite nucleosides with limited range of dye options. Alternatively,

    monomers bearing appropriate functional groups can be incorporated into

    oligonucleotides and subsequently reacted with functionalised labels amine-

    NHS ester coupling, for example. This approach is constrained by compatibility

    with the phosphoramidite method and labelling reaction efficiencies.

    A synthetic strategy is reported that offers an efficient route to multiple

    labelling or modification of an oligonucleotide. Orthogonal protecting group

    strategies enable solid phase conjugation to phosphoramidite derivatives of label

    molecules. Five modified nucleoside phosphoramidites have been synthesised

    and incorporated into DNA oligonucleotides. Modifications at the 2- and 5-

    position of pyrimidine nucleosides with Fmoc, Levulinyl and TBDMS protecting

    groups have been investigated. A HyBeacon probe, multiply labelled with FAM

    was synthesised and melting studies indicated successful discrimination between

    mutant and wild-type target sequences, with biophysical results very close toconventional probe technologies. Advantages of this strategy include

    compatibility with the phosphoramidite method, practical convenience, broad

    scope of label options and high-yielding label installation.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    4/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    5/140

    Contents

    ABSTRACT' I!

    ACKNOWLEDGEMENTS' 3!

    ABBREVIATIONS'&'ACRONYMS' 4!

    INTRODUCTION' 1!

    Nucleic'Acid'Structure5' 2!Nucleosides!&!Nucleotides! 2!Watson1Crick!Base!Pairing!Specificity! 4!Monomers!to!Oligos! 5!

    Oligonucleotides' 7!Oligonucleotide!Synthesis! 7!The!Phosphoramidite!Monomer! 9!The!Phosphoramidite!Cycle! 0!

    Oligonucleotides'in'Genetic'Analysis' 18!

    HyBeacon'Probes' 19!

    Labelling'Strategies' 21!Previous!Strategies!for!Multiple!Labelling!of!Oligonucleotides! 25!

    A!New!Strategy!for!Adding!Multiple!Reporter!Groups!to!Oligonucleotides! 3!

    Retrosynthesis'&'Alternative'Approaches' 34!

    RESULTS'&'DISCUSSION' 37!

    Monomer'Design' 37!

    Synthetic'Approach' 41!51O1(4,41Dimethoxytrityl)121O1(21(91fluorenylmethyloxycarbonyl)ethyl)151methyluridine131 O1

    (O121cyanoethyl1N,N1diisopropyl)!phosphoramidite!(Monomer!)! 4!51O1(4,41Dimethoxytrityl)151(31(91fluorenylmethyloxycarbonyl)1propynyl)121deoxyuridine131 O1

    (O121cyanoethyl1N,N1diisopropyl)!phosphoramidite!(Monomer!2)! 46!51O1(4,41Dimethoxytrityl)121O1(61(tert1butyldimethylsilyloxy)hexanamido)ethyl)151

    methyluridine131O1(O121cyanoethyl1N,N1diisopropyl)!phosphoramidite!(Monomer!3)! 49!51O1(4,41Dimethoxytrityl)151(31(61(tert1butyldimethylsilyloxy)1hexanamido)1propynyl)121

    deoxyuridine131O1(O121cyanoethyl1N,N1diisopropyl)!phosphoramidite!(Monomer!4)! 55!51O1(4,41Dimethoxytrityl)151(31(61(levulinyloxy)1hexanamido)1propynyl)121deoxyuridine131 O1

    (O121cyanoethyl1N,N1diisopropyl)!phosphoramidite!(Monomer!5)! 56!51O1(4,41Dimethoxytrityl)151(31(61(91fluorenylmethyloxycarbonyl)1hexanamido)1propynyl)121

    deoxyuridine131O1(O121cyanoethyl1N,N1diisopropyl)!phosphoramidite!(Monomer!6)! 57!

    Oligonucleotide'Synthesis'Strategy' 59!Approach!A! 59!

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    6/140

    Approach!B! 6!Approach!C! 66!HyBeacon!Studies! 67!

    CONCLUSION'&'FUTURE'WORK' 71!

    Conclusion' 71!

    Future'Work' 72!Variant!Labelling! 72!Branching!Oligonucleotides! 73!Cell!Imaging! 74!Combinatorial!Labelling! 74!

    EXPERIMENTAL' 77!

    General'Methods' 77!General! 77!Spectroscopic! 77!Phosphitylation! 78!The!Phosphoramidite!Method! 78!Oligonucleotide!Purification!&!Characterisation! 79!Melting!Analysis! 79!

    Synthesis'of'Modified'Monomers' 80!MONOMER!! 80!MONOMER!2! 85!MONOMER!3! 88!MONOMER!4! 92!MONOMER!5! 94!

    REFERENCES' 99!

    APPENDICES' 1!

    Interim'Report' 1!Introduction! !Results!&!Discussion! 4!Proposals!for!Future!Work! 7!References! 0!

    Oligonucleotide'Synthesis'Optimisation' 11!Conditions! !

    Mass'Spectrometry'Results' 12!HPLC1MS!&!Deconvoluted!Peaks! 4!

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    7/140

    Acknowledgements

    Thank you to Professor Tom Brown for taking account of my interests and

    personal ambitions and assigning me a project that suited them so well. To be

    given a project with real significance in the progression of the field is an honour

    and knowing that my work is of some importance has been a great motivator.

    I would very much like to thank Dr. Nittaya Gale for working closely with

    me on this project and for making it both enjoyable and the success that it has

    been. I have learnt the rewards of persistence in problem solving and I have also

    learnt to remain positive when faced with disappointment or mistakes an asset

    to a healthy work-attitude that extends beyond the laboratory. I have been

    privileged to work alongside someone who has demonstrated and passed on

    such a high standard of laboratory skills and practices.

    Thank you to Dr. Simon Gerrard for always taking the time to answer my

    questions. Thank you to Xiaomei Ren for submitting many spectroscopic samples

    on my behalf. Thank you to Dr. Afaf El-Sagheer for on-hand advice, for keeping

    an eye on me and for making me go home when its getting late. Working

    alongside such hard-working, exceptional people has been inspiring.

    For their love, support and interest in all that I do, thank you to my Mum

    and Dad.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    8/140

    Abbreviations & Acronyms

    A adenine

    angstrom(s)

    Ac acetyl

    AcOH Acetic acid

    Act activating group

    aq. aqueous

    bp base pair(s)

    br broad (spectral)

    Bu n-Bu normal (primary) butyl

    tBu tert-butylBz benzoyl

    C cytosine

    C degrees Celsius

    cat catalytic

    CEP-(Cl) 2-Cyanoethyl N,N-diisopropyl(chloro) -

    phosphoramidite

    cm centimetre(s)

    conc. concentrated

    COSY correlation spectroscopy

    CPG controlled pore glass

    chemical shift in parts per million downfield from

    tetramethylsilane

    d day(s); doublet (spectral); deci; deoxy

    DCA dichloroacetic acid

    DCC N,N-dicyclohexylcarbodiimideDCM dichloromethane

    DEA diethylamine

    DEPT distortionless enhancement bypolarisation transfer

    DIC diisopropylcarbodiimide

    DIPEA diisopropylamine (Hnigs base)

    DMAP 4-(N,N-dimethylamino)pyridine

    DMF dimethylformamide

    DMSO dimethylsulfoxide

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    9/140

    DMTr 4,4-dimethoxyltrityl

    DNA 2-deoxyribonucleic acid

    ds double-stranded

    E1CB

    unimolecular (conjugate base) elimination

    EDC 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide

    EDTA ethylenediaminetetraacetic acid

    eq. equivalent

    ESI electrospray ionisation

    Et ethyl

    Et2O diethyl ether

    EtOAc ethyl acetate

    EtOH ethanolFAM fluorescein amidite

    FCC flash column chromatography

    FISH fluorescence in-situhybridisation

    Fmoc 9-fluorenylmethoxycarbonyl

    FT-ICR Fourier transform ion cyclotron resonance

    G guanine

    g gram(s)

    h hour(s)

    HPLC high-performance liquid chromatography

    HRMS high-resolution mass spectrometry

    Hz hertz

    iB iso-butyl

    iPrOH iso-propanol

    J coupling constant (in NMR spectrometry)

    K Kelvin(s) (absolute temperature)

    L litre, dm3

    Lev levulinyl

    LPE light petroleum ether

    wavelength

    lit. literature value (abbreviation used with full stop)

    LRMS low-resolution mass spectrometry

    micro

    m multiplet (spectral); metre(s); milliM molar (moles per litre); mega

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    10/140

    M+ parent molecular ion

    Me methyl

    MeCN acetonitrile

    MeOH methanol

    MHz megahertz

    min minute(s)

    mM millimolar (millimoles per litre)

    mol mole(s); molecular (as in mol wt)

    MS mass spectrometry

    MW

    mol. wt. molecular weight

    m/z mass-to-charge ratio

    n normalNHS N-hydroxysuccinimide

    NMI N-methylimidazole

    nm nanometre(s)

    NMR nuclear magnetic resonance

    NTP nucleoside triphosphate

    OD optical density

    ONT oligonucleotide

    PAGE polyacrylamide gel electrophoresis

    PG protecting group

    Ph phenyl

    pip piperidine

    ppm part(s) per million

    Pr propyl

    iPr isopropyl

    PS polystyrene

    py pyridine

    q quartet (spectral)

    Rep reporter group

    Rf

    retention factor (in chromatography)

    RNA ribonucleic acid

    RP-HPLC reverse phase high performance liquid

    chromatography

    rs residue (gene, protein)rt room temperature

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    11/140

    s singlet (spectral); second(s)

    SN1 unimolecular nucleophilic substitution

    SN2 bimolecular nucleophilic substitution

    SNP singlenucleotide polymorphism

    STR short tandem repeats

    Su succinimide

    t triplet (spectral)

    t time; temperature in units of degrees Celsius (C)

    T absolute temperature in units of Kelvins (K); thymine

    Tac2O tertbutylphenoxyacetyl acetic anhydride

    TAMRA carboxytetramethylrhodamine

    TBAF tetrabutylammonium fluorideTBDMS tert-butyldimethylsilyl chloride

    TCA trichloroacetic acid

    TEA triethylamine

    TEAA tetraammonium acetate

    tert tertiary

    TFA trifluoroacetic acid

    THF tetrahydrofuran

    TLC thin-layer chromatography

    TM

    melting temperature

    TMS trimethylsilyl; tetramethylsilane

    TR

    retention time

    UV ultraviolet

    vis visible

    vol volume

    v/v volume per unit volume (volume-to-volume ratio)

    wt. weight

    w/w weight per unit weight (weight-to-weight ratio)

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    12/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    13/140

    Introduction

    DNA is a biological information storage molecule that contains the genetic

    code a sequence made from an alphabet consisting of only four letters (A, G, C

    & T), yet holds the fundamental instructions for growth and proliferation of

    almost all living systems.3 With entire complex life forms operating on such

    fundamentally simple instructions, it is not surprising that small errors in this

    code can have serious consequences. Diagnostic techniques that interrogate the

    genetic code are invaluable tools for forensic and medical science.4

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    14/140

    Nucleic Acid Structure5

    Nucleosides & Nucleotides

    DNA is a biological macromolecule (figure 1). In nature, it exists

    predominantly in the double stranded DNA double helix form. Each strand of the

    helix is a polymer of nucleotides, figure 6, of which there are four possibilities,

    dependent on which of the four heterocyclic bases they contain (figure 2).

    Figure 1 B-DNA -The Iconic Double Stranded Helix2

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    15/140

    Figure 2 Purines and pyrimidines - the four heterocyclic bases of DNA, nucleoside & nucleotide

    structure and atom numbering convention

    The 2-deoxyterminology indicates the absence of a hydroxyl group at the

    2-position of the ribose sugar, the main structural differentiation between DNA

    and ribonucleic acids (RNA), which has a hydroxyl group at the 2-position. The

    other distinguishing feature between DNA and RNA is the replacement of the

    pyrimidine thymine (T), DNA, with uracil (U), RNA (figure 3).

    Figure 3 Distinctions in heterocyclic bases in DNA/RNA

    N

    NN

    N

    NH2

    NH

    NN

    N

    O

    NH2

    N

    N

    NH2

    O

    NH

    N

    O

    O

    O

    OH

    ROBase

    Base =(A) (G) (C) (T)

    Purine Pyrimidine

    R = H, Nucleoside

    R = PO32-, Nucleotide

    1'

    2'3'

    4'

    5'

    1

    2

    3

    4

    5

    67

    1

    2

    3

    4

    5

    68

    9

    A = Adenine

    G = Guanine

    C = Cytosine

    T = Thymine

    Heterocyclic

    NH

    N

    O

    O

    NH

    N

    O

    O

    Uracil (RNA) Thymine (DNA)

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    16/140

    Watson-Crick Base Pairing Specificity

    The two antiparallel strands of the DNA duplex are said to be

    complementary in their pairing. Each base hydrogen bonds specifically to its

    complementary, or opposite, partner. The two strands are bound primarily by

    this specific hydrogen-bonding pattern and the overall structure is additionally

    stabilised by other non-covalent interactions such as base-stacking and entropic

    contributions from the internally-positioned hydrophobic bases. It is the specific

    hydrogen bonding between bases, however, that confers information storage

    ability.

    Figure 4 Watson-Crick Base Pair Hydrogen Bonding Scheme

    James Watson and Francis Crick elucidated the structure of the DNA double

    helix in 1953 with the help of the work of several other prominent researchers.

    The base-pairing scheme (figure 4) was named in their honour. Watson-Crick

    base pairing specificity is the basis upon which DNA can accurately copy itself.

    During semi-conservative DNA replication, the helix is separated and the twoseparated halves of the parent provide all the information required for the

    formation of two exact-copy daughter strands (figure 5).

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    17/140

    Figure 5 The semi-conservative nature of DNA replication, a consequence of Watson-Crick base

    pairing specificity2

    The key concept is that DNA is an accurate and consistent source of

    biological information.

    Monomers to Oligos

    Polymeric nucleoside strands are connected through a 3,5-

    phosphodiester bond (figure 6) giving DNA/RNA strands intrinsic directionality.

    At physiological pH (7.4), the phosphate groups exist in their deprotonated,

    anionic form, hence the name - nucleic acid. The two strands of a double helix

    run in opposite directions to each other, termed antiparallel.

    Parent Strand

    Daughter Strands

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    18/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    19/140

    Figure 7 Three nucleic acid duplex conformations, A, B & Z. B is the most common2

    Oligo, from Greek few, is the prefix attached to nucleotides to describe

    polymeric single nucleic acid strands from anywhere between a dinucleotide

    (dimer) to several hundred mers. Oligonucleotides will bind readily to form

    stable duplexes with complementary sequences. It is this key aspect of the

    oligonucleotide that makes it a perfect tool for investigating specific DNA

    sequences, a tool with applications in diverse fields due to the significance of the

    information that DNA contains.

    Oligonucleotides

    Oligonucleotide Synthesis

    The chemical synthesis of oligonucleotides follows a solid-phase strategy

    conceived in 1959 by Bruce Merrifield. His invention that was originally designed

    for the task of peptide synthesis was awarded the 1984 Nobel Prize in Chemistry.

    Application of the solid-phase approach to oligonucleotide synthesis retains all

    the advantages of the Nobel Prize-winning conception.6

    The phosphoramiditemethod has opened new doors in hybrid DNA research, stimulating scientific

    A B Z

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    20/140

    progress in biochemistry, molecular biology, pharmacology, drug design &

    development and gene technology.

    Synthesis on a solid matrix enables synthesis of nucleotide polymer

    lengths of up to around 200 - far longer than would otherwise be possible.1b

    Complex purifications are replaced by quick, simple and efficient washes that

    regularly achieve stepwise yields in excess of 99.5% - a prerequisite for the

    viability of longer oligonucleotide syntheses. Large reagent excesses can be

    applied, driving reactions rapidly to completion. The nature of the process is

    well-suited to computerised automation, enabling highly efficient and

    reproducible syntheses.

    Table 1 Impact of coupling efficiencies on overall yield

    Length

    of Oligo

    Average Stepwise Yield (Coupling Efficiency)

    90% 95% 97% 98.5% 99.5%

    10 38.7 63.0 76.0 87.3 95.6

    FinalYield(

    %)20 13.5 37.7 56.1 75.0 90.9

    50 - 8.1 22.5 47.7 78.2

    100 - - 4.9 22.4 60.9

    150 - - 1.1 10.5 47.4

    200 - - - 4.9 36.9

    As demonstrated in table 1, high-yielding coupling reactions are necessary

    for the successful synthesis of oligonucleotides of reasonable length.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    21/140

    The Phosphoramidite Monomer

    The key aspect of nucleoside building blocks is the phosphoramidite

    moiety at the 3-alcohol (figure 8), which undergoes reaction with exposed 5-

    alcohol groups under mild acid catalysis. The DMTr group that protects the 5-

    alcohol is also of importance as it prevents off-target polymerisation of the

    monomers.

    Figure 8 Highlighted features of a dT phosphoramidite monomer

    HN

    O

    O N

    O

    O

    O

    PO

    NN

    O

    O

    Dimethoxytrityl moiety

    2-Cyanoethyl N,N-diisopropylphosphoramidite moiety

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    22/140

    The Phosphoramidite Cycle

    Oligonucleotide synthesis via the phosphoramidite method begins with a

    single protected nucleoside that is covalently attached to a solid support, often

    polystyrene or controlled-pore glass (CPG) granules 500-3000 in diameter,

    within the confines of a column. Elongation of the nucleotide chain occurs

    through the sequential addition of phosphoramidite monomers by a series of

    chemical processes that constitute the phosphoramidite cycle (figure 10).

    Figure 9 Diagram of solid-phase oligonucleotide column

    The functionalised resin column (figure 9) is loaded onto the DNA

    synthesiser and subjected to the synthesis cycle.

    Solvents & reagent solutionspass through freely

    Filters retain growingoligonucleotides onsolid supports

    Insoluble solid support(resin) particles anchoroligonucleotides withinthe column

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    23/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    24/140

    Table 2 A typical DNA oligonucleotide synthesis cycle for a 0.2 mol scale synthesis

    Operation Reagent/Solvent Time/s

    Wash Acetonitrile 30

    Detritylate 3% TCA/DCM 50

    Monitor trityl - -

    Wash Acetonitrile 30

    Flush Argon 10

    Couple0.1 M phosphoramidite monomer and 0.5 M tetrazole in

    acetonitrile30

    Wash Acetonitrile 30

    Flush Argon 10

    CapAcetic anhydride/py/THF 1/1/8 and 17.6% w/v N-methyl

    imidazole in acetonitrile30

    Wash Acetonitrile 30

    Flush Argon 10

    Oxidise 0.02 M iodine in water/pyridine/THF 2/20/78 45

    Wash Acetonitrile 30

    Flush Argon 10

    Detritylation

    The resin is functionalised with protected nucleoside monomers. The

    DMTr protecting group prevents polymerisation during functionalisation, but

    must be removed to allow coupling to the next base (figure 11). The acid-

    catalysed detritylation is achieved with a 3% TCA/DCM or 3% DCA/DCM solution.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    25/140

    Figure 11 Acid-catalysed (TCA or DCA) detritylation mechanism

    The trityl cation is a highly-conjugated and stable species, absorbing

    strongly in the visible wavelength range. Automatic and real-time UV-Vis

    absorption analysis with a radiation source (495 nm

    ) of the column effluent

    quantifies the efficiency of the synthesis, terminating it if yields fall below a

    certain threshold to preserve reagents.7

    Activation & Coupling

    Coupling to the next base (figure 12) requires a mild acid catalyst,

    tetrazole (or derivative of), to activate the phosphorous of the incoming

    nucleoside phosphoramidite group towards nucleophilic attack. The protonation

    of the diisopropylamino group makes it a better leaving group and facilitates

    O

    O

    O

    O

    O

    BH2

    O

    O

    NH

    O

    OCl

    Cl

    ClH

    O

    O

    O

    O

    O

    BH2

    O

    O

    NH

    O

    OCl

    Cl

    Cl

    H

    O

    O

    HO

    O

    O

    NH

    BH2

    O

    O

    O

    O

    etc.

    DMT cation (orange)

    = Resin (CPG, PS)

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    26/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    27/140

    5-alcohol groups to prevent further participation of erroneous oligonucleotides

    in further elongation cycles (figure 13).

    Figure 13 An electrophilic acylation cocktail prevents deletion-mutation oligonucleotide inclusion

    in further elongation cycles

    The reaction of acetic anhydride with NMI creates a strong nucleophile

    and also an equivalent of acetic acid. Pyridine is therefore included in the

    capping cocktail to maintain the alkaline conditions required to avoid premature

    detritylation.

    Oxidation

    The product of the coupling reaction yields a phosphorous (III) linkage

    between the bases. This phosphite triester must undergo oxidation to an acid-

    stable phosphorous (V) phosphotriester before the next acidic detritylation takesplace (figure 14). Treatment with iodine in water and pyridine affords the

    cyanoethyl-protected PV backbone.

    O

    O

    HOB

    O

    OHN

    NN

    O O

    O

    O

    NN

    O

    O

    OB

    O

    OHN

    O

    HO

    ON

    NN

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    28/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    29/140

    Figure 15 Ester hydrolysis of succinyl linker - cleavage from solid support

    Once cleaved from the solid support, the oligonucleotide/conc. aq.

    ammonia solution is heated at 55C for 5 h in order to remove the phosphorous

    backbone-protecting cyanoethyl groups and any exocyclic nucleobase nitrogen-

    protecting groups (figure 16), however, the choice of protecting group varies

    depending on the modification within the oligonucleotide e.g. those unstable to

    standard conditions.8

    Figure 16 Standard protection of exocyclic amines for phosphoramidite oligonucleotide synthesis

    The oligonucleotide can then be purified by the method appropriate to the

    oligonucleotide and application e.g. gel filtration, HPLC (reverse phase or ion

    exchange-HPLC) or PAGE.

    O

    O

    OB

    O

    OHNNH4

    OH

    O

    OHN

    OH

    O

    OH

    OB+/- H

    NH3

    N

    HN

    ON

    O

    OH

    HO

    O

    PhNH

    N

    N

    O

    NHN

    O

    OH

    HO

    O

    N(4)-benzoyl dC (dCBz) N(2)-isobutyryl dG (dGiB)

    N

    NN

    N

    HN

    O

    OH

    HO

    Ph

    O

    N(1)-benzoyl dA (dABz)

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    30/140

    Oligonucleotides in Genetic Analysis

    Watson-Crick base pairing specificity makes short

    oligonucleotides ideal as reliable and sequence-specific probes of

    complementary sequences. Labelling of such oligonucleotides is

    required, as an oligonucleotide possesses no inherent

    detectability. Labelling probes with fluorescent organic molecules

    (dyes) confers many useful properties which are exploited in the

    various synthetic oligonucleotide applications.1b Many different probe

    technologies have been developed, a common theme being the generation of a

    fluorescent signal upon hybridisation to complementary sequences.9

    Figure 17 Components of a FAM dye phosphoramidite used for labelling ONTs at 5 terminus

    The labelling of these probes is achieved through covalent linkage

    between the oligonucleotide and the corresponding functionalised derivatives of

    the labels. Fluorescent dyes, quenchers and affinity tags with pre-installed

    coupling functionality (figure 17) are commercially available and their properties

    are well-documented.10 Many applications, such as HyBeacon probes, require,

    or can be enhanced by, the inclusion of multiple fluorescent labels.1b

    O

    O

    O

    O

    ONH

    O

    O

    O

    OP

    O

    N

    N

    Reactivegroup

    Spacer Signalling moiety

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    31/140

    HyBeacon Probes

    An excellent application and measure of success of the fluorescent

    labelled oligonucleotide is the HyBeacon probe. HyBeacons are linear single-

    strand oligonucleotides labelled with fluorescent groups, such as fluorescein.

    HyBeacons are the simplest format of oligonucleotides used in genetic analysis,

    for example in probing single nucleotide polymorphisms (SNP)s and short

    tandem repeats (STRs).4a The principal of the HyBeacon probe is to produce an

    increase in fluorescence upon hybridisation to a complementary sequence (figure

    18). This fluorescent enhancement upon hybridisation occurs due to the

    reduction of distance-dependent fluorescence quenching interactions between

    fluorophores and between fluorophores and nucleobases.11

    Upon hybridisation,the formation of the ordered and relatively rigid helical duplex reduces the

    proximity between fluorophores and quenching structures reliably increasing

    fluorescence.12

    Figure 18 Signal generation principle of a HyBeacon probe

    Correctly-paired Watson-Crick bases contribute to a more

    thermodynamically stable duplex than one containing mismatches.13 The

    analytical technique exploiting this property is the melt study. The melting

    temperature (TM) is the temperature at which the two complementary strands

    make the transition between the separate, free strands and the hybridised

    duplex. Raising the temperature of a duplex eventually provides enough thermal

    energy to overcome the inter-strand attractive interactions and the duplex

    denatures; upon cooling the process is reversed. Duplex formation/denaturation

    is tracked by changes in fluorescence; the TM

    is taken as the point of inflection

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    32/140

    on a fluorescence vs. temperature plot maxima of the first derivative (figure

    19).

    Figure 19 Melt curve and first derivative of a oligonucleotide duplex, TM= 53C, with correct

    Watson-Crick base pairing

    Melting experiments quantify duplex stability and can accurately

    differentiate between perfect and imperfectly paired gene sequences.

    Experiments using HyBeacon probes provide a quick and cheap means of

    detecting SNPs, STRs and mutations through fluorescent measurement.

    -50

    0

    50

    100

    150

    200

    250

    300

    350

    400

    450

    7,000

    8,000

    9,000

    10,000

    11,000

    12,000

    35 37 39 41 43 45 47 49 51 53 55 57 59 61 63 65 67 69 71

    -dFI/

    dT

    /K-1

    Fluoresc

    entIntensityat

    525

    (FI)

    /

    ArbitraryUnits

    Temperature (T) / C

    Melt Curve and First Derivative of anOligonucleotide Duplex

    Raw Melt Data

    First Derivative

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    33/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    34/140

    Labelling is most commonly achieved through phosphoramidite ligation,

    a), by the inclusion of a modified monomer containing the functional moieties or

    linker arms described that react with the incoming phosphoramidite label.15

    Phosphoramidite labelling is advantageous in that it is high-yielding and

    compatible with the automation of the DNA synthesiser.

    Each type of labelling reaction has its strengths and limitations: Methods

    b, c, and d) are post-synthetic techniques requiring amino or thiol-modified

    monomers and corresponding electrophilic label derivatives. They offer a

    strategy for introducing labels that arent stable to oligonucleotide deprotection

    conditions. They may suffer from low chemoselectivity due to reactivity of the

    electrophilic labels with amino groups of the oligonucleotide bases.

    16

    Additionally, the corresponding resultant functional group connections are

    susceptible to hydrolysis, an outcome only partially avoidable via careful

    handling of pH with appropriate buffer system.16 They are not the most efficient

    strategies.

    Enzymatic incorporation of labelled NTPs, strategy e), is limited to small

    scales, but does allow for high-density multiple label incorporation.16 Due to

    enzyme-substrate specificity, however the nature of the dye is limited.16 Other

    conceivable labelling strategies include the Staudinger ligation and Diels-Alder

    type reactions (figure 21).16

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    35/140

    Figure 21 Labelling by h) & i) Diels-Alder [4+2] cycloadditions and j) Staudinger ligation

    Compatibility with solid-phase synthesis; retention of dsDNA helix

    stability; retention of Watson-Crick pairing specificity and practical synthetic

    convenience are challenges in the area of oligonucleotide labelling. Certain in

    vivoapplications add the demands of bioorthogonality and toxicity to this list.17

    The properties of a particular label must be considered carefully to assess

    suitability. The labelling process must be compatible with oligonucleotide

    chemistry i.e. easy to attach with mild reagents. Its mode of attachment mustretain the required properties of the label e.g. fluorescence. Interference with

    oligonucleotide function, i.e. specific hybridisation and duplex formation, should

    be minimal. Additionally, the stability of the labelled oligonucleotide should be

    sufficient to withstand storage at -20C.

    Multiple labelling is often desirable. Detection of complementary

    sequences is often limited by the concentrations at which the targets are

    present. Low concentrations yield low intensity fluorescence upon hybridisation.

    LOligo NL

    Oligo

    h)

    i)

    O

    O

    O

    N

    O

    O

    O

    LOligo NL

    Oligoj)

    O

    N

    O

    O

    O

    LN3Oligo

    OligoO

    O

    Ph2P

    O

    O

    NH

    Ph2P

    O

    L

    O

    L Oligo= Label with Spacer, = Oligonucleotide

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    36/140

    Although highly sensitive equipment exists, detection of target sequences

    at biological concentrations i.e. picomolar or lower, can prove challenging.18

    Enhancing the fluorescent signal of a probe through addition of multiple

    fluorescent reported groups helps address this problem. Fluorescence in-situ

    hybridisation (FISH) is a technique for imaging living cells and visualising the loci

    genes in a chromosome. FISH typically uses 100-1000 bp probes with a labelling

    frequency of 1 fluorescent label per 30 nucleotides. For greater specificity in

    probe-binding, shorted sequences are desirable, but limitations come with the

    low levels of fluorescence this produces. FISH probes are typically synthesised by

    enzymatic incorporation of fluorescently-labelled nucleotide triphosphates in a

    polymerase process such as nick translation, random priming or the polymerase

    chain reaction.

    9a

    Enzymatic incorporation of fluorescent nucleotides, however,limits the diversity of fluorescent groups that can be attached due to enzyme-

    substrate specificity.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    37/140

    Previous Strategies for Multiple Labelling of Oligonucleotides

    Currently, the most common strategy for multiple labelling is via the

    inclusion of commercially-available phosphoramidite monomers with pre-

    installed dye functionality, such as the monomer shown in figure 22. Prominent

    companies offering such compounds include Glen Research8, Link Technologies19

    and Invitrogen20.

    Figure 22 Components of a general reporting phosphoramidite monomer, such as thosecommercially available from Glen Research8

    Such dye phosphoramidite monomers are, however, often expensive and

    they are limited by the requirement that the dye and conjugating functional

    groups (amide bonds in the above example) must withstand the oligonucleotide

    synthesis cycles.

    Another current approach is through the inclusion of monomers bearing

    the functional groups outlined in figure 22. Amide coupling yields, however,

    rarely match those attainable by phosphoramidite coupling. A commercially

    available amino modifier is shown in figure 23.

    O

    O

    O

    N

    HN

    O

    O

    O

    NH

    HN

    O

    O

    O

    P O

    NN

    Rep

    Phosphoramidite nucleobase Linker Reporter group(Rep)

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    38/140

    Figure 23 Fmoc Amino-Modifier C6 dT, 5'-(4,4-Dimethoxytrityl)-5-[N-((9-fluorenylmethoxycarbonyl)-aminohexyl)-3-acrylimido]-2'-deoxyUridine,3'-[(2-cyanoethyl)-( N,N-diisopropyl)]-phosphoramidite, commercially available from suppliers, such as Glen Research8

    Incorporation of monomers bearing orthogonal protecting groups into the

    oligonucleotide is less well-documented, but offers several advantages over the

    previously mentioned approached. Post-synthetic deprotection and exposure of

    alcohol groups permits coupling to dye phosphoramidites that do not require the

    same levels of resilience to the oligonucleotide phosphoramidite synthesis cycle.

    This broadens the scope of potential dyes/labels/reporter groups and may offer

    a cheaper and high-yielding multiple labelling strategy.

    Previous methods, using modified, protected phosphoramidite monomers,

    have achieved successful internal labelling (figure 24).21 These strategies typically

    involve interruption of the DNA oligonucleotide solid-phase synthesis at the

    point of modification addition. This is then allows manual deprotection of the

    modification, and labelling with phosphoramidite-functionalised fluorescent

    dyes, such as TAMRA or FAM, before returning to the DNA synthesiser to

    complete the sequence.

    O

    O

    O

    N

    HN

    O

    O

    O

    N

    H

    HN

    O

    O

    O

    O

    P O

    NN

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    39/140

    Figure 24 Internal labelling monomer used in previous work by Dobson et al. 2-Fmoc uridineCEP, 5-O-(4,4-Dimethoxytrityl)-2-O-[2-(9-fluorenylmethyloxycarbonyloxy)]ethyl uridine

    The oligonucleotide synthesis strategy for the incorporation and labelling

    of such monomers is outlined in figure 25.

    O

    O

    O

    N

    NH

    O

    O

    O

    O

    PO

    NN

    OO O

    O

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    40/140

    Figure 25 Previous synthetic approaches to internal labelling21

    The implication of this strategy is that, in order to achieve multiple

    labelling, the automated DNA synthesis cycle must be interrupted at the point of

    3'5'

    OPG

    3'5'

    OH

    Couple to reporter phosphoramidite

    3'5'

    3'5'

    Cleavage & exocyclicnucleobase deprotection

    3'5'

    Extend

    Extend

    Remove, deprotect

    Return to synthesiser forextension until sequence complete

    PG = Protecting Group

    Modified Monomer

    Solid Support (PS/CPG)

    Rep = Reporter Molecule

    PO

    O

    O

    O

    Rep

    PO

    O

    O

    O

    Rep

    PO

    O

    O

    O

    Rep

    5'

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    41/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    42/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    43/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    44/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    45/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    46/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    47/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    48/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    49/140

    Results & Discussion

    Monomer Design

    For most applications, the success of a particular oligomer modification is

    judged primarily by two factors: signal sensitivity upon hybridisation and

    retention of normal hybridisation behaviour. A consequence of the highly

    sophisticated and precise nature of the DNA double helix is that random

    modifications to its components are more likely to interfere with, rather than

    improve duplex stability and Watson-Crick complementarity.

    Modification at 2-position does not directly interfere with the

    phosphoramidite synthesis method, but steric bulk may reduce coupling

    efficiencies.22 Distancing of bulky groups from the 3-CEP group with alkyl

    spacers is expected to reduce steric inhibition of the coupling reaction.

    Modification at the C-5 position of pyrimidines is a common target since it

    is not involved base-pair hydrogen bonding. The modification is projected into

    the major groove of the helix, allowing for significant steric tolerance and

    minimal inhibition of helix formation (figure 33).

    Figure 33 Modification at pyrimidine C-5 projects into major groove

    N

    NN

    NN

    NN

    O

    O

    HH

    H NN

    N

    N O

    N

    NN

    N

    O

    H

    HH

    H

    H

    Major

    Groove

    Minor

    Groove

    Major

    Groove

    Minor

    Groove

    = Modification

    A T G C

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    50/140

    The protecting groups chosen must be orthogonal to the conditions used

    to remove the DMTr group (acid labile). The fluorenylmethoxycarbonyl (Fmoc)

    moiety was used as an alcohol-protecting group, introduced by nucleophilic acyl

    substitution and removed by -elimination (figure 34). -elimination is easily

    achieved with mild bases, such as morpholine, piperidine, piperazine, for

    example, due to the electron-withdrawing fluorene ring system. Decarboxylation

    also provides a strong thermodynamic driving force.

    Figure 34 Fmoc cleavage by (1,2)-elimination with piperidine

    The 2-deoxyuridine version of monomer 1 has been used previously by

    Dobson et al. in the successful synthesis of a dual-labelled probe using Fmoc

    chemistry.15 This monomer is now synthesised via an improved syntheticstrategy. Use of this monomer to construct a multiply-labelled oligonucleotide

    RO O

    OH

    RO O

    O

    14 e , 4n+ 2, n= 3Planar Aromatic System

    HN

    NHH

    NH

    RO

    O

    O

    NHH

    R OH

    - CO2

    N

    - eliminationE1cb

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    51/140

    was attempted, but afforded disappointing yields indicating the need for

    optimisation of the synthesis protocol.

    The second generation of protecting groups were levulinyl and TBDMS.

    The levulinyl protecting group was introduced vianucleophilic acyl substitution

    from the corresponding acid anhydride. Hydrazinolytic cleavage was achieved

    with hydrazine monohydrate in pyridine/acetic acid (figure 35).

    Figure 35 Post-synthetic removal of levulinyl protecting group by hydrazinolysis

    The tert-butyldimethylsilyl (TBDMS) protecting group was introduced by

    nucleophilic substitution (SN1) and removed with fluoride chemistry, usingTEA.HF as fluoride source (figure 36).

    Figure 36 TBDMS removal viahypervalent silyl intermediate

    TBDMS and levulinyl (Lev) protecting groups were selected as suitable

    protecting groups due to their proven orthogonality with DNA oligonucleotide

    synthesis, as demonstrated in a previous use in the construction of cross-linked

    DNA.23

    O

    O

    O

    H2N NH2

    +/- HOH

    O

    OHN

    H2N

    OSi

    F

    Si OFSi

    F

    HO

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    52/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    53/140

    Synthetic Approach

    5-O-(4,4-Dimethoxytrityl)-2-O-(2-(9-fluorenylmethyloxycarbonyl)ethyl)-5-

    methyluridine-3-O-(O-2-cyanoethyl-N,N-diisopropyl) phosphoramidite (Monomer 1)

    Precursor 9 was synthesised in accordance with the method described by

    Richardson et al, 2009. The first transformation in this sequence, (i), was

    achieved with 79% yield. This compares well with the reported literature figure of

    90% when the relative scales of synthesis are considered.9e

    NH

    O

    ON

    O

    OHOH

    HO

    N

    O

    O N

    O

    OH

    HO(i)

    N

    O

    O N

    O

    OH

    O(ii)

    NH

    O

    ON

    O

    OOH

    O (iv)

    NH

    O

    ON

    O

    OOH

    O (v)

    NH

    O

    ON

    O

    OO

    O

    DMTr

    DMTr

    OHO

    FmocCEP O

    Fmoc

    DMTr DMTr

    (iii)

    1109

    7 8

    Scheme 1 Synthetic route to Fmoc-protected monomer 1 (i) Diphenyl carbonate (1.1 eq.), NaHCO3

    (cat), DMF, 100C, 18 h, 79%; (ii) DMT-Cl (1.4 eq.), py, rt, 24 h, 67%; (iii) Ti(i

    PrOH)4 (1.4 eq.), ethyleneglycol (5.0 eq.), DMF, rt, 22 h, 62%; (iv) Fmoc-Cl (1.1 eq.), DIPEA (2.0 eq.), DCM, rt, 2.5 h, 40%; (v)CEP-Cl (1.2 eq.), DIPEA (2.0 eq.), DCM, rt, 3 h, 71%.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    54/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    55/140

    The second transformation, the protection of the 5-alcohol with the trityl

    group viaa nucleophilic substitution (SN1) reaction with dimethoxytrityl chloride

    is both mechanistically and practically simple. Regioselectivity for the 5-alcohol

    in preference to the 3-alcohol was accomplished on grounds of steric hindrance.

    The DMTr-Cl is a bulky tertiary alkyl halide that does not react readily with the

    secondary alcohols at either 2- or 3-positions. The substitution reaction

    proceeds with the concomitant loss of hydrogen chloride from the reagents

    necessitating the presence of at least one equivalent of a suitable base due to

    the acid-ability of the resulting DMTr protecting group. The reactivity of the

    tertiary DMTr carbocation with to the hydroxide ion calls for careful exclusion of

    water. The necessarily basic conditions of this reaction mean that the presence

    of water would lead to rapid undesired consumption of DMTr-Cl throughformation of DMTr-OH.

    The reaction was achieved in 67% yield, lower than that reported in the

    literature value- 89%.9e The smaller scale predisposed the reaction to a lower

    yield, but other factors also contributed. The reaction proved to be stubborn and

    additional aliquots of DMTr-Cl, time and patience were required. Eventually (t =

    22 h), the reaction was determined incomplete, but was quenched, worked-up

    and purified regardless. Upon repetition of this reaction, more rigorous

    exclusion of water would be observed.

    A notable feature of syntheses whose reagents or products include the

    DMTr protecting group is the distinctive orange colour that indicates presence of

    the DMTr cation in solution (figure 38). This can alert the chemist to the

    presence of moisture in solvents, for example. Due to the strong molar

    extinction coefficient of the DMTr cation however, it is rare that reaction

    mixtures remain completely colourless as very low levels of the cation confer

    colouration. Also, heat-induced detritylation of compounds offers a rapid and

    informative TLC plate visualisation technique.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    56/140

    Figure 38 UV Absorption spectra of DMT cation in acetonitrile24

    Opening of the anhydro ring (figure 39) was achieved in a 62% yield,

    slightly lower than the literature value of 79%.9e A possible cause for disparities

    in apparent yield success may be due to the contamination of the product

    reported in the literature with titanium complex remains accounting for higherapparent yield. Thorough, repeated centrifugation of the reaction mixture

    ensured that a highly pure product was obtained, but inevitably at the loss of

    some compound to the retentate, although this was washed several times and

    checked viaTLC until satisfactorily clean.

    Figure 39 Nucleophilic alkoxide ring opening mechanism

    Absorption, /nm

    400 450 500 550 600

    0.0

    0.5

    1.0

    1.5

    498 70 000 dm3 cm-1 mol-1

    410 28 960 dm3 cm-1 mol-1

    Absorbance(unitless)/ArbitraryUnits

    Ti(OiPr)4HO

    OH

    N

    O

    O N

    O

    OH

    ODMTr

    Ti(OCH2CH2O)O

    O

    H

    HN

    O

    O N

    O

    OH

    ODMTr

    TiL3

    O

    O

    +/- H

    +/- L

    HN

    O

    O N

    O

    OH

    ODMTr

    O

    OH

    2 Ti(OCH2CH2O)2

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    57/140

    The major source of loss was due to compound-impurity overlap during

    column chromatographic purification that necessitated the exclusion of impure

    fractions from the pure fraction pool. Total recovery of compound with a second

    column could be achieved if it were needed for subsequent syntheses, or if a

    valuable amount of reagents had been committed and warranted isolation. A

    competitive reaction that may have reduced yield involves the isopropoxide

    acting as the ring-opening nucleophile. However, due to greater steric bulk of

    the isopropoxide and using an excess of ethylene glycol, this pathway does not

    appear to interfere significantly.

    Fluorenylmethyloxycarbonyl was chosen to protect the remaining primary

    alcohol group as it offers orthogonal compatibility with the conditions of thephosphoramidite method (see experimental, the phosphoramidite method).

    Nucleophilic substitution of chloride at the chlorocarbonate group of Fmoc-Cl by

    the primary alcohol yields the product with the concomitant overall loss of

    hydrogen chloride from the reagents. The basic conditions employed in the

    reaction protect the DMT-protected alcohol group from acid-induced exposure

    and deprotonation of the alcohol assists in its nucleophilic attack at the Fmoc

    carbonyl carbon atom.

    The reaction was initially attempted using pyridine functioning as both

    solvent and base. Bis-addition of Fmoc resulted, as determined by LRMS.

    Experimentation with reaction conditions led to the use of DIPEA as base and

    DCM as solvent, addition of Fmoc-Cl as solution and at 0C. This had three

    advantages. Firstly, the reaction proceeded more reliably and required fewer, if

    any, extra additions of Fmoc-Cl. Secondly, addition of Fmoc-Cl as a solution is

    practically cleaner than exposing the reaction to the external environment and

    allows for more control over addition rate. Thirdly, DCM can be removed quicker

    and more completely than pyridine, which was often detectable by NMR in early

    experiments. The combination of lower reaction temperatures and very gradual

    addition of Fmoc-Cl solution successfully afforded the desired product, 40%.

    Further optimisation would benefit this reaction.

    The phosphitylation was performed with great care and afforded the

    phosphoramidite monomer 1 in 71% yield. High product purity was required, asno further purification would take place before the compound entered the

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    58/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    59/140

    Figure 40 The Palladium & Copper mediated catalytic cycles of the Sonogashira reaction26

    The product was only obtained in 49% yield. Possible side products

    include the alkyne-alkyne homocoupled dimer and the furano-pyrimidine

    nucleoside (figure 41).

    ThePalladiumCycle

    TheCopperCycle

    OxidativeAddition

    trans-cisIsomerisation

    Reductive

    Elimination

    RR' R' X

    Transmetallation

    RCu

    Cu X

    RH

    Cu X

    R3N

    R3NH X

    PdII

    L

    L

    R' X

    PdIIR'

    L

    L

    R

    RCu

    PdII

    R'

    L

    L R

    PdL2

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    60/140

    Figure 41 Proposed furano-pyrimidine side-product formation mechanism

    Fmoc protection was successful, achieving a yield of 82%. Originally, this

    was attempted using the less-reactive reagent - Fmoc-OSu to achieve the

    required primary alcohol selectivity, but the reaction did not proceed so gradual

    addition and low temperature, 0C, was employed.27

    Phosphitylation to yield monomer 2 was moderately successful as

    reflected in the yield of 60%. Improvements in yield could be made through

    obtaining better separation of crude components. The product eluted over 18fractions indicating that a lower-polarity eluent system would not be suitable to

    achieve better separation of components. Instead a larger volume, wider

    diameter column could be tried in order to achieve separation with a slightly

    more polar system.

    N

    O

    O N

    O

    OH

    O

    OH

    DMTr

    Et3N H

    Cu

    N

    O

    O N

    O

    OH

    O

    OH

    DMTrEt3N

    Cu

    H

    N

    O

    O N

    O

    OH

    ODMTr

    OH

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    61/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    62/140

    The mechanisms of hydrazinolysis and the Staudinger reduction are both

    particularly interesting and are shown in figures 42 and 43, respectively.

    Figure 42 Mechanism of primary amine deprotection by hydrazinolytic phthalimide cleavage

    The phthalimide-protected amine can be installed via an SN2 reaction

    between the corresponding primary alkyl halide and potassium phthalimide.29

    The combined subsequent phthalimide deprotection constitutes the Gabriel

    synthesis.29 This precursor was synthesised in-house by Dr. Montserrat

    Shelbourne, following the approach described by Manoharan et al.30

    NH

    O

    ON

    O

    OOH

    ODMTr

    N

    NH

    O

    ON

    O

    OOH

    ODMTr

    NH2

    O

    ONH2

    NH2NH

    O

    ON

    O

    OOH

    ODMTr

    NH

    O

    O

    HNH2N

    +/- H

    +/- H

    14

    O

    HN

    HN

    O

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    63/140

    Figure 43 Primary amine formation mechanism by Staudinger reduction, affording primary amine,

    14 upon aqueous work-up

    Nucleophilic attack by triphenylphosphine at the terminal azide nitrogen

    followed by ring closure forms a phosphazide. Irreversible loss of nitrogen forms

    the iminophosphorane. Hydrolysis occurs upon aqueous work-up and isthermodynamically driven by the formation of the strong phosphorous-oxygen

    double bond.

    To extend the length of the linker at the 2-amino ethoxy dT, an amide

    coupling between the NHS-ester and the amine was used to give compound 15 in

    78% yield. The activated acid offers good reactivity; the reaction proceeded

    rapidly without need for carbodiimide coupling agents. Amide coupling is a

    frequently-employed connection approach throughout the syntheses of these

    monomers. A general mechanism is given in figure 44.

    NH

    O

    ON

    O

    OOH

    ODMTr

    NH214

    NH

    O

    ON

    O

    OOH

    ODMTr

    N

    N

    N PPh3

    NNN

    PPh3

    N

    NN

    PPh3

    N PPh3 N PPh3-N2

    H2O

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    64/140

    Figure 44 General mechanism of amide bond formation

    NHS-activated acids offer enhanced reactivity, permitting the use of milder

    conditions, by providing a more stable leaving group than the hydroxide anion.

    They are of intermediate reactivity - between that of carboxylic acids and acid

    chlorides. They are easy to handle yet their enhanced reactivity is still significant

    enough to offer a real synthetic advantage. Occasionally the amide couplingdoes not proceed satisfactorily and carbodiimides can be used to drive the

    reaction. The mechanism of this process is illustrated in figure 45.

    Figure 45 Carbodiimide-promoted amide bond formation

    Undesired side-reactions can occur from the O-acylisourea and include a

    rearrangement for form a stable N-acylurea and a reaction with an additional

    carboxylic acid molecule to form an acid anhydride of the carboxylic acid (figure

    46).

    R1 NH2

    HO

    O

    R2 +/- H

    - H2O

    NH

    O

    R2

    R1

    NC

    NG G

    NH

    CN

    G G

    O

    O

    RH

    O

    O

    R

    G NH

    N

    O

    G

    O R NH2R'

    +/- H

    R'NH

    O

    R NH

    O

    NH

    GG

    O-acylisourea

    G =

    cyclohexyl (DCC)

    isopropyl (DIC)ethyl (EDC)

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    65/140

    Figure 46 O-acylisourea rearrangement forming stable N-acylurea

    The acid anhydride can go on to react with the amine to form the desired

    amide and release a carboxylic acid (figure 47).

    Figure 47 Reaction of carboxylic acid with O-acylisourea to form dicyclohexylurea and acid

    anhydride - further reacting to give desired amide and an equivalent of corresponding carboxylic

    acid

    Phosphitylation was successful and without complication. Column

    purification however, was problematic. Elution was protracted and caused

    overlap with excess phosphitylating reagent. The pure fractions yielded only

    29%.

    G

    HN N

    O

    G

    O

    R N-acylurea

    NH

    O

    N

    O

    R

    G

    G

    G =cyclohexyl (DCC)isopropyl (DIC)ethyl (EDC)

    O

    O

    R

    G NH

    N

    O

    G

    O R+/- H

    R'NH

    O

    R

    N

    H

    O

    N

    H

    GG

    * Acid anhydrideof carboxylic acid

    R

    O

    O

    O

    R

    H2N R'

    *

    R

    O

    OH G =cyclohexyl (DCC)isopropyl (DIC)ethyl (EDC)

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    66/140

    A common problem is the inclusion of unreacted phosphitylating reagent.

    When elution of this reagent, or if PV species overlap with that of the monomer,

    the contaminated fraction(s) cannot be combined. Their presence can be

    visualised on TLC plates stained with p-anisaldehyde and are observed as a white

    spot. This is easily distinguishable from the monomer that stains black with p-

    anisaldehyde. If this separation proved particularly difficult for a particular

    compound then the reaction could be performed with phosphitylating reagent in

    limiting proportions.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    67/140

    5-O-(4,4-Dimethoxytrityl)-5-(3-(6-( tert-butyldimethylsilyloxy)-hexanamido)-propynyl)-

    2-deoxyuridine-3-O-(O-2-cyanoethyl-N,N-diisopropyl) phosphoramidite (Monomer 4)

    The TBDMS group was installed viaunimolecular nucleophilic substitution

    (SN1) of its chloride derivative. The acid was activated to the NHS ester to

    promote amide coupling. The NHS group is a better leaving group than the

    hydroxide anion and coupling can be achieved with milder conditions.

    Amide coupling via the NHS-activated acid proceeded without

    complication. The clean reaction facilitated simple column chromatography and

    afforded compound 16 in 63% yield. Phosphitylation was achieved in 70% yield.

    HN

    O

    O N

    O

    O

    O

    NH

    4CEP

    O

    O

    TBDMS5

    DMTr

    HN

    O

    O N

    O

    OH

    O

    N

    H

    O

    O

    TBDMS5

    DMTr

    OH

    O

    HO

    HN

    O

    O N

    O

    OH

    O

    NH2

    DMTr

    OH

    O

    OTBDMS

    O

    O

    OTBDMS

    N

    O

    O

    (i)

    (ii)

    (iii)

    (iv)

    16

    Scheme 4) Synthetic route to TBDMS-protected monomer 4 (i) TBDMS-Cl (1.2 eq.), DIPEA (2.0 eq.),DCM, rt; (ii) NHS (1.3 eq.), EDC-Cl (1.2 eq.), DIPEA (3.0 eq.) DMF, rt; (iii) DIPEA, (1.5 eq.), DCM, DMF, rt,4 h 63% iv CEP-Cl 1.2 e . DIPEA 2.0 e . DCM rt 2.5 h 70%.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    68/140

    5-O-(4,4-Dimethoxytrityl)-5-(3-(6-(levulinyloxy)-hexanamido)-propynyl)-2-

    deoxyuridine-3-O-(O-2-cyanoethyl-N,N-diisopropyl) phosphoramidite (Monomer 5)

    Compound 17 was produced by a carbodiimide-assisted amide formation

    reaction. Synthesis of compound 18 was achieved in 70% yield through reaction

    between the alcohol and the acid anhydride, which was prepared in-situ. This

    terminal alkyne product was subsequently coupled to the 5-iodouridine by a

    Sonogashira cross-coupling reaction in 59% yield.

    HN

    O

    O N

    O

    O

    O

    NH

    5CEP

    O

    O

    Lev5

    DMTr

    HNO

    O N

    O

    OH

    O

    NH

    O

    O

    Lev5

    DMTr

    OH

    O

    HO

    NH

    O

    HO

    (i)

    HN

    O

    O N

    O

    OH

    ODMTr

    I

    NH

    O

    OLev

    (iv)

    (iii)

    17 18

    19

    O

    O

    OH

    O

    O

    O

    O

    O

    (ii)

    (v)

    Scheme 5 Synthetic route to levulinyl-protected monomer 5 (i) Propargylamine (1.5 eq.), EDC-Cl(1.5 eq.), DMF, rt, 18 h, 67%; (ii) DCC, Et2O, rt; (iii) Levulinic anhydride (prepared in-situ) (1.4 eq.),DMAP (0.5 eq.), DCM, DMF, rt, 1.5 h, 70%; (iv) CopperI Iodide (0.25 eq.), Pd0(PPh3)4 (0.1 eq.), DMF, TEA,rt, 1.5 h, 59%; (v) CEP-Cl (1.2 eq.), DIPEA (2.0 eq.), DCM, rt, 3.5 h, 74%.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    69/140

    5-O-(4,4-Dimethoxytrityl)-5-(3-(6-(9-fluorenylmethyloxycarbonyl)-hexanamido)-

    propynyl)-2-deoxyuridine-3-O-(O-2-cyanoethyl-N,N-diisopropyl) phosphoramidite

    (Monomer 6)

    Scheme 6 (i) TMS-Cl (1.2 eq), DIPEA (2.0 eq.), DCM, rt, 3 h, 64%; (ii) Fmoc-Cl (1.2 eq.), DIPEA (2.0

    eq.), DCM, rt; (iii) EDC-Cl (1.3 eq.), DIPEA (3.0 eq.), DCM, rt, 2 h, 56%; (iv) Py.HF (1.1 eq.), py, DCM,

    unsuccessful; (v) Not yet attempted

    Inclusion of a TMS protection step improved a previous approach.

    Previously the Fmoc-protected acid was reacted with compound 20 without the

    3-O-TMS protecting group and resulted in bis Fmoc coupling. Deprotection of

    the TMS group, however, resulted immediate cleavage of the DMTr group. An

    attempt to recover the compound was made since the DMTr group could be

    selectively reinstalled at the 5-position, however the recouped yield was

    insufficient to warrant further efforts.

    Approaching monomer 6 via the 5-O-(4,4-Dimethoxytrityl)-5-(3-(6-

    (hydroxy)-hexanamido)-propynyl)-2-deoxy uridine, treated with Fmoc-O-

    succinimide did not proceed even with addition of 4-dimethylaminopyridine, acoupling catalyst (figure 48).

    HN

    O

    O N

    O

    O

    O

    NH

    CEP

    O

    O

    Fmoc5

    DMTr

    HN

    O

    O N

    O

    OH

    O

    NH

    O

    O

    Fmoc5

    DMTr

    HN

    O

    O N

    O

    O

    O

    NH

    O

    O

    Fmoc5

    DMTr

    TMS

    HN

    O

    O N

    O

    O

    O

    NH2

    DMTr

    HN

    O

    O N

    O

    OH

    O

    NH2

    DMTr

    (i)

    (iv) (v)

    TMS

    O

    OHHO

    O

    OHO

    Fmoc

    (ii)

    (iii)

    20 21

    22 6

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    70/140

    Figure 48 Mechanism of catalytic action of DMAP in an amide coupling reaction

    The reaction mixture was worked-up and dried in vacuoand the reaction

    attempted at -42C (MeCN, CO2(s)) with the more reactive Fmoc reagent Fmoc

    chloride. The reaction produced four DMTr-containing products as identified by

    TLC, but which degraded to starting material before purification was achieved. It

    is suspected that the presence of pyridine or remaining DIPEA from the reaction

    mixture, which was added to maintain basicity during the HCl-producingreaction, was responsible for the premature Fmoc cleavage. In conclusion,

    handing of compounds bearing the Fmoc moiety must be treated carefully with

    respect to over-exposure to basic conditions.

    N

    N

    OO

    O

    N

    O

    O

    N

    N

    O

    O

    N

    N

    O

    O

    O

    OO

    O

    N N

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    71/140

    Oligonucleotide Synthesis Strategy

    Modified monomers 1-5 were introduced into a HyBeacon probe by three

    different approaches. Following deprotection of the modifications, the exposed

    groups were labelled with 6-FAM phosphoramidite. The probe sequence was

    programmed to differentiate between wild-type and mutant gene for an SNP

    implicated in statin-induced myopathy, rs4149056.31

    Approach A

    In the first approach (figure 49), the modified monomer is incorporated

    into the sequence at the desired positions as follows. The sequence is

    synthesised until after the coupling of the modified monomer. Keeping the

    terminal 5- DMTr group on, the synthesis is paused and the oligonucleotide

    column is treated with an appropriate cocktail to remove protecting group (Lev,

    Fmoc or TBDMS) of the hydroxyl. The column returned to the synthesiser and the

    synthesis cycle resumed at the point of coupling to the next nucleoside in the

    sequence. The label phosphoramidite is coupled to the modified monomer and

    then the sequence continued until, at the next modification, the process isrepeated until the sequence is complete.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    72/140

    Figure 49 Labelled-oligonucleotide synthesis approach A

    3'5'

    OPG

    Remove, deprotect

    3'5'

    OH

    Couple to 6-FAM-phosphoramidite

    3'5'

    Return to synthesiser for

    extension until next modification

    3'5'

    Cleavage & exocyclic nucleobase deprotection

    3'5'

    Extend

    Extend

    Return to synthesiser forextension until sequence complete

    3'5'

    PG = Protecting Group

    Modified Monomer

    Solid Support (PS/CPG)

    Extend

    Rep = Reporter Molecule, FAM

    PO O

    O

    ORep

    PG

    O

    PO OO

    ORep

    PO O

    O

    O

    Rep

    PO O

    O

    O

    Rep

    PO O

    O

    ORep

    PO O

    O

    ORep

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    73/140

    This approach was tested with monomer 1 with the following sequence, where

    superscript XF represents FAM labelling:

    5-GTGGATA1FATGCG1FTCATGG

    The crude HPLC-MS and the MS are shown in figure 50. The major peak

    corresponds to the correct oligonucleotide product and indicates a yield of

    around 80%. This approach afforded a high-purity probe (figure 50), but the

    complexity of the procedure may deter many from its use.

    The labelled oligonucleotide product was isolated and tested as a

    HyBeacon probe in a melting analysis.

    Figure 50 Left:HPLC trace for monomer 1, sequence: GTGGATA1FATGCG1FTCATGG (1F = monomer1, labelled with FAM) synthesised with Approach A, correct ONT product (2 FAM additions), T

    r: 8.6

    min; Right: MS showing the correct expected product calc. MS = 7535.97 Da, found MS =7536.41Da)

    Approach B

    Approach B (figure 51) offers a less-complicated labelling protocol for the

    installation of multiple labels. The entire oligonucleotide is synthesised with the

    protected, modified bases in their desired location within the sequence, the

    terminal 5-DMTr is left on. The oligonucleotide column is removed from the

    synthesiser, treated with DEA (as above), the modification protecting groups

    removed and the column returned to the synthesiser at the point of next

    coupling. Finally, the phosphoramidite label is applied and the synthesis is

    complete, ready for standard deprotection and cleavage.

    4 5 6 7 8 9 10 11 Time [min]

    0

    50

    100

    150

    Intens.

    [mAU]

    res3210_GB5_01_14311.d: UV Chromatogram, 290nm

    4511.6 4949.05314.1

    5650.0

    6317.2

    6767.4

    7206.4

    7536.4

    9028.2

    -MS, 8.5-8.7min, Deconvoluted (MaxEnt)

    0

    1

    2

    3

    4x10

    Intens.

    4000 5000 6000 7000 8000 9000 10000 11000 m/z

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    74/140

    Figure 51 Labelled-oligonucleotide synthesis approach B

    Several oligonucleotides were synthesised following this approach in order

    to optimise the conditions. For monomers 1 and 2, the results were

    unsatisfactory (figures 52 & 53) 0-15% product was found. Attempts at

    optimisation were made, changing capping conditions from acetic anhydride to

    Tac2O (milder capping reagent) and no capping. Further optimisation attempts

    5'

    Remove, deprotect

    Cleavage & exocyclic nucleobase deprotection

    3'5'

    Synthesise entire ONT

    PG = Protecting Group

    Modified Monomer

    Solid Support (PS/CPG)

    Rep = Reporter Molecule, FAM

    3'5'

    OPGOPG

    Couple to 6-FAM-phosphoramidite

    3'

    OHOH

    5' 3'

    O

    5'

    5'

    P

    OO

    O

    Rep

    O

    P

    OO

    O

    Rep

    O

    POO

    O

    Rep

    O

    POO

    O

    Rep

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    75/140

    included doubling the coupling times (10 to 20 mins), concentration of the

    applied phosphoramidite monomer (0.1 to 0.2 M) and using fast-deprotecting

    nucleosides (dGDMF, dCAc). An example HPLC trace is shown in figure 52, full

    details are given in HPLC section of the appendix.

    Figure 52 Left: HPLC trace for res3261, sequence: GTGGATA2FATGCG2FTCATGG3 (2F = monomer 2,labelled with FAM, 3 = propanol), synthesised with Approach B, peak at T

    R= 7.4 min: no FAM, 8.1

    min: only 1 FAM; Right: Mass spec of peak at TR

    = 8.1 min

    Figure 53 HPLC UV 290 nm

    chromatrogram for res3263, sequence GTGGATA1FATGCG1FTCATGG3 (1F= monomer 1, labelled with FAM, 3 = propanol), peak at T

    R= 7.4 min no FAM, 8.1 min: only 1 FAM

    addition and TR

    = 8.6 min: expected product with 2 FAM label. Right: MS of peak at TR= 8.6 min.

    Monomer 4 was incorporated into the following sequence (res3373):

    5-GTGGATA4FATGCG4FTCATGG

    Deprotection of monomer 3 was attempted with TEA (20% in MeCN, 45

    min) then TEA.3HF (2 x (0.3 mL, 30 min)). The mass spectrometry results did not

    produce any clear peaks (figure 54). Purification was attempted with RP-HPLC,but no separation was achieved. This could be due to the TEA.3HF, a strong

    0 2 4 6 8 10 12 14 16 Time [min]

    0

    50

    100

    150

    200

    Intens.

    [mAU]

    Res3261c_RC1_01_14667.d:UV Chromatogram,290 nm

    6325.26421.2

    6629.2

    6958.3

    -MS,7.9-8.1min,Deconvoluted(MaxEnt)

    0

    1

    2

    3

    4

    4x10

    Intens.

    5500 5750 6000 6250 6500 6750 7000 7250 7500 7750 8000 m/z

    0 2 4 6 8 10 12 14 16 Time [min]

    0

    50

    100

    150

    200

    Intens.

    [mAU]

    Res3263c_RC3_01_14669.d: UV Chromatogram, 290 nm

    5394.06011.1

    7535.4

    9007.6 9420.4 10528.8 13192.5

    -MS, 8.6-8.7min, Deconvoluted(MaxEnt)

    0.00

    0.25

    0.50

    0.75

    1.00

    1.25

    4x10

    Intens.

    5000 6000 7000 8000 9000 10000 11000 12000 13000 14000 m/z

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    76/140

    base, prematurely deprotecting the exocyclic nucleobase amine groups causing

    FAM to add to free amines of the nucleobases, resulting in numerous products

    and preventing successful purification. Optimisation is required.

    Figure 54 HPLC UV 290 nm

    chromatogram for res3373, using polystyrene support (TBDMSdeprotection incompatible with CPG, fluoride ion reacts with the Si-O structure of the CPG)

    Monomer 5 was incorporated into the following sequence:

    GTGGATA5F

    ATGCG5F

    TCATGG

    Table 3 Experimentation with ONT synthesis

    ONT ID Conditions Chromatogram

    res3371

    CPG resin

    Treated with 20% DEA/MeCN, 25 min to deprotect

    the acetal capping of the CPG hydroxyl groups

    A

    res3390

    CPG resin

    No treatment with DEA (control experiment) Bres3391 PS resin C

    All oligonucleotides in table 3 were treated with DEA (10% DEA/MeCN, 10

    min) to deprotect the phosphate backbone prior to levulinyl deprotection.

    Levulinyl deprotection conditions were hydrazine monohydrate:acetic

    acid:pyridine, 0.031:0.5:2.0 v/v/v, rt, 12 min.

    0 2 4 6 8 10 12 14 16 Time [min]

    0

    50

    100

    150

    Intens.

    [mAU]

    RES3373_RC4_01_16570.d: UV Chromatogram, 290 nm

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    77/140

    A

    B

    C

    Figure 55 HPLC UV 290 nm

    chromatograms, A) res3371, B) res3390 & C) res 3391 sequence:GTGGATA5FATGCG5FTCATGG (5F = monomer 5, labelled with FAM), peak at T

    R= 7.5 min:

    unlabelled; TR= 8.2 min: 1 FAM addition; T

    R= 8.9 min: 2 FAM additions (correct product); MS traces

    are shown in appendix (HPLC and Deconvoluted MS)

    Treatment with DEA prior to synthesis produces a higher amount of

    product than when untreated (figure 55). It is suspected that coupling to FAM

    phosphoramidite was reduced by migrating acetyl groups from the CPG-hydroxy

    protection. These were removed with the DEA and were therefore not present to

    complicate capping.

    0 2 4 6 8 10 12 14 16 Time [min]

    0

    10

    20

    30

    40

    Intens.

    [mAU]

    RES3371_RC5_01_16571.d: UV Chromatogram, 290 nm

    0 2 4 6 8 10 12 14 16 Time [min]

    0

    20

    40

    60

    Intens.

    [mAU]

    RES3390_RC6_01_16572.d: UV Chromatogram, 290 nm

    0 2 4 6 8 10 12 14 16 Time [min]

    0

    10

    20

    30

    Intens.[mAU]

    RES3391_RC7_01_16573.d: UV Chromatogram, 290 nm

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    78/140

    Approach C

    A further improvement in terms of practical simplicity was developed in

    approach C (figure 56). Interruption of the synthesis at the point of labelling was

    avoided. The terminal DMTr group at the end of synthesis was removed (DMTrOFF) and the 5-alcohol capped with hex-5-ynyl phosphoramidite (X). This

    means that no human intervention is required during the synthesis and a

    standard phosphoramidite cycle program can be used without further

    manipulation.

    Figure 56 Labelled-oligonucleotide synthesis approach C

    5'

    Remove, deprotect

    Cleavage & exocyclic nucleobase deprotection

    3'5'

    Synthesise entire ONT (DMTr OFF), cap with hex-5-ynyl phosphoramidite ('X')

    PG = Protecting Group

    Modified Monomer

    Solid Support (PS/CPG)

    Rep = Reporter Molecule, FAM

    3'5'

    OPGOPG

    Couple to 6-FAM-phosphoramidite

    3'

    OHOH

    5' 3'

    O

    5'

    5'

    P

    OO

    O

    Rep

    O

    P

    OO

    O

    Rep

    OP

    OO

    O

    Rep

    OP

    OO

    O

    Rep

    X

    X

    X

    X

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    79/140

    This strategy was used to synthesis the following oligonucleotide

    (res3415) with monomer 5 on a PS solid support:

    5-XGTGGATA5FATGCG5FTCATGG

    Figure 57 Left: HPLC UV 290 nm

    chromatogram for res3255, sequence: GTGGATA5FATGCG5FTCATGG(5F = monomer 5, labelled with FAM), peak at T

    R= 8.6 min: [M-134], unknown, peak at T

    R= 8.8 min:

    2 FAM additions (correct product); Right: MS

    This approach yielded the correct product (figure 57) in a high-yield and

    was accomplished with a practically-uncomplicated protocol. This represents a

    successful new strategy for adding multiple reported groups to oligonucleotides.

    The dual-labelled oligonucleotide was tested as a HyBeacon probe.

    HyBeacon Studies

    A melting study provided a direct comparison between a conventionally-

    labelled HyBeacon and one synthesised by the new strategy (figure 58). A

    HyBeacon was synthesised using fluorescein dT, purchased from Glen

    Research. The HyBeacon probes were identical apart from the mode of

    fluorescein attachment, permitting an assessment of the effect of the phosphate

    linker on fluorescence and duplex stability.

    0 2 4 6 8 10 12 14 16 Time [min]

    0

    200

    400

    600

    800

    1000

    Intens.

    [mAU]

    RES3415CR1_RB3_01_17066.d:UV Chromatogram,290nm

    7206.2

    7743.3

    -MS,8.7-9.0min,Deconvoluted (MaxEnt)

    0.0

    0.5

    1.0

    1.5

    5x10

    Intens.

    5000 5500 6000 6500 7000 7500 8000 8500 m/z

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    80/140

    Figure 58 Structural comparison of FAM dT monomers: A commercially-available, = F in res1137

    and B - as produced by the reported strategy (5F,used in res3391)

    The probe sequence was programmed as the complement for an SNP at

    rs4149056, implicated in Statin-induced myopathy, forming a G:C base pair in

    the matched sequences (WT) and G:A in the mismatched. The melting

    temperatures of the probe-target duplexes were investigated. The results are

    displayed in table 3.

    Table 4 Melting temperature results of HyBeacon(TM) probe comparison study

    Oligo.ID SequenceDuplex T / C TM / CWild Type Mutant

    Probe 1res1137 GTGGATATAFGCGFTCATGG 43.0 53.0 10.0

    Probe 2res3210 GTGGATATA5GCG1TCATGG 42.0 52.0 10.0

    Probe 3res3391 GTGGATATA5GCG5TCATGG 45.0 54.0 9.0

    WTTarget

    res3375 CCATGAACACATATATCCAC --

    MTTarget

    res3376 CCATGAACGCATATATCCAC --

    O

    O

    O

    N

    HN

    O

    O

    O

    NH

    HN

    O

    O

    O

    HO

    O

    OH

    O

    O

    O

    N

    HN

    O

    O

    NH

    O

    OPO

    O O

    NH

    O

    O

    O

    HO

    O

    OH

    A

    B

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    81/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    82/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    83/140

    Conclusion & Future Work

    Conclusion

    This project set out to investigate a new strategy for adding multiple

    reporter groups to oligonucleotides through incorporation of protected modified

    monomers. Five monomers were synthesised and incorporated into

    oligonucleotides. The most successful were tested as HyBeacon probes. The

    probes successfully differentiated between the two alleles of a gene implicated in

    statin-induced myopathy. The properties of the probe-target duplexes with

    probes synthesised by the reported strategy and a probe synthesised using a

    FAM-dT-phosphoramidite, purchased from Glen Research8, were compared. The

    probes were found to have high similarity, indicating that the structural

    differences in the base-label connection do not significantly interfere with either

    duplex stability or fluorescent emission. This was a successful demonstration of

    the strategy. It is clear that an effective multiple labelling strategy has been

    developed.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    84/140

    Future Work

    Variant Labelling

    Theoretically, through the use of two or more modified monomers in

    which the label-linking functional groups (-OH or NH2) are mutually orthogonal

    in their protection, multiple and variant labels could be easily installed (figure

    61).

    Figure 61 Capacity for multiple and variant labelling

    5'

    Remove, deprotect PGA

    Cleavage & exocyclic nucleobase deprotection

    3'5'

    O(Rep)AO(Rep)A O(Rep)B O(Rep)B

    3'

    PG = Protecting Group

    Modified Monomer

    Solid Support (PS/CPG)

    Rep = Reporter Molecule

    3'5'

    OPGAOPGB OPGBOPGA

    Couple to reporter phosphoramidite A

    3'

    OHOPGB OPGBOH

    5'

    O(Rep)AO(Rep)A O(Rep)B O(Rep)B

    5'3'

    O(Rep)AOPGB OPGBO(Rep)A

    Deprotect PGB

    5' 3'

    O(Rep)AOH OHO(Rep)A

    Couple to reporter phosphoramidite B

    Synthesise Entire ONT

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    85/140

    Variant label installation could prove useful in applications that desire

    dendrimeric structures, asymmetric branching, or installation of multi-coloured

    probes for wider instrument detection compatibility.

    Branching Oligonucleotides

    Installing multiple reporter groups could enhance HyBeacon probe

    resolution. Ultra-high fluorescence outputs could be achieved through the use of

    branching side chains containing multiple fluorescent groups (figure 62).

    Similarly, fluorescence in-situ hybridisation (FISH) target detection capability

    could be improved through higher fluorophore installation levels.

    Figure 62 A) Orthogonally protected phosphoramidite monomer, B) Reporter groups or branchextensions C) Branched oligonucleotide

    DNA and RNA sequences can be detected by surface-enhanced Raman

    spectroscopy (SERS).32 The probe sequence is usually connected through a

    thioether at the 3-terminus to a gold nanoparticle and labelled with a Raman-

    active dye. The anchored strand has many degrees of freedom and the proximity

    of the Raman dye to the gold surface, required for Raman enhancement, cannot

    HN

    O

    O N

    O

    O

    O

    CEP

    DMTr

    O

    OPG2

    OPG1HN

    O

    O N

    O

    O

    O

    DNA

    DNA

    O

    O

    O

    Rep1 or ONTs

    Rep2 or ONTs

    Rep1 or ONTs

    Rep2 or ONTs

    Rep3 or ONTs

    Rep4 or ONTs

    Rep5 or ONTs

    Rep6 or ONTs

    A) B)

    C)

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    86/140

    be guaranteed. Branching oligonucleotides could provide multiple anchor points,

    maintaining dye-surface proximity and optimum Raman signal enhancement.

    Cell Imaging

    For imaging sequences in living cells, the probe must enter the cell. This

    can be achieved manually by microinjection or with by addition of a delivery

    vehicle, such as lipofectamine, but both techniques can damage the cell and alter

    its normal functioning. Natural cellular uptake mechanisms can be encouraged to

    accept molecules bearing polyamines (figure 63), which exist in their protonated

    cationic form at physiological pH.

    33

    A route for simple ligation of the sperminegroup and fluorescent groups to oligonucleotides may be of use in cell imaging

    research.

    Figure 63 Spermine phosphoramidite, commercially available from Glen Research

    Combinatorial Labelling

    The reported strategy for adding multiple reporter groups to

    oligonucleotides is particularly well suited to large-scale synthesis. This is due to

    the simple labelling protocol and single purification step. It is time and cost-

    efficient, essential for viability of commercial applications.

    N N N NO

    TFA TFA

    TFA TFA

    O P O

    NN

    O

    O

    TFA = trifluoroacetyl protecting group F

    FF

    O

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    87/140

    Simultaneous, detection of many specific pathogens, bacterial, viral,

    fungal, parasitic, could be achieved with a cocktail of FISH probes, each with

    specific pathogen targets. The synthesis of the probe cocktail could be easily

    accomplished with a combinatorial process. Complete probe sequences can be

    combined on their solid supports then deprotected and labelled in one batch.

    The probe cocktail could be tailored to its application. A hand-wash gel,

    loaded with the correct probe sequences could simultaneously detect the

    predominant bacterial pathogens responsible for hospital infections or provide a

    rapid detection system for biological warfare agents for use in high-risk facilities.

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    88/140

  • 7/27/2019 A Strategy for Adding Multiple Reporter Groups to Oligonucleotides by Tom Fleming

    89/140

    Experimental

    General Methods

    General

    Standard reagents were procured from Aldrich, Avocado or Fluka. Solvents

    were distilled as follows: DCM, TEA, DIPEA, py from CaH2

    and THF from sodium

    with benzophenone as indication of anaerobic, anhydrous quality. Reaction