20160_ftp

Embed Size (px)

DESCRIPTION

7

Citation preview

  • 5/23/2018 20160_ftp

    1/7

    The anthracycline antibiotics:antitumor drugs that alterchromatin structureAzra Rabbani,1 Ron M. Finn,2 and Juan Ausio2*

    SummaryAnthracycline antibiotics are an important group ofantitumor drugs widely used in cancer chemotherapy.However, despite the increasing interest in these che-motherapeutic agents, their mechanism of action is notyet completely understood. Here, we review what iscurrently known about the molecular mechanisms in-volved with special emphasis on the interaction of thesedrugs with chromatin and its constitutive components:DNA and histones. The evidence suggests that one veryimportant component of the activity of these drugs is

    the result of these manifold interactions that lead to achromatin unfolding and aggregation. This chromatinstructural disruption is likely to interfere with the meta-bolic processes of DNA (replication and transcription)and it may play an important role in the apoptosis under-gone by the cells upon treatment with these drugs.BioEssays27:50 56, 2005. 2004 Wiley Periodicals, Inc.

    Introduction

    Although anthracycline antibiotics were recognized for their

    antibacterial properties in 1939, their chemical characteri-

    zation experienced a substantial increase only after the

    therapeutic value of their antitumor activity was described in

    the early 1960s. Two of these compounds daunomycin

    (daunorubicin) and adriamycin (hydroxyl daunomycin or

    doxorubicin) were initially isolated fromStreptomyces peuce-

    tius varcaesitue(1,2) and exhibit the widest spectrum of

    antitumor activity against human cancers.(3,4) Because of the

    increasing potential medical relevance of these drugs in cancer

    chemotherapy, there has been a lot of interest in chemically

    modifying them to try to minimize their cardiotoxic side

    effects.(4,5) In this regard, many anthracycline derivatives

    have been produced in recent years.(6,7) A good example can

    be found withpixantrone {6,9-bis[(2-aminoethyl) amino] benzo

    (g) isoquinoline-5, 10-dione dimaleate}, which has already

    gone through phase II clinical trials and which shows a lot of

    promise for the treatment of Non-Hodgkins lymphoma.(8)

    Daunomycin andadriamycinwitha chemicalformulaof C27H29 NO10 andC27 H29 NO11, respectively, are soluble in water,

    ethanol and methanol. They are planar molecules consisting

    of a tetracycline ring bound to a daunosamine through a

    glycoside bond. The quinone and hydroquinone groups ofthese molecules act as electron donors and acceptors.(9)

    Anthracyclineantibiotics and their derivatives are an integral

    component of the current chemotherapy in the treatment of

    solid tumors and leukemia; indeed daunomycin has beenused

    primarily to treat adult myelogenous leukemia.(10,11) Although

    there is still some controversy about their mode of action,

    the anticancer activity or cytotoxic effects may involve the

    interaction with nuclear components, especially DNA and type

    II topoisomerases;they mayalso participate in theinhibition of

    DNA and RNA synthesis.(9,1214)

    Characterization of the binding sites of antitumor drugs in

    the cell is important because it can provide clues required for

    further drug design to enhance drug efficacy. This reviewfocuses its attention on progress made on the understanding

    of the interaction of anthracycline antibiotics adriamycin and

    daunomycin with different chromatin components.

    Interaction with DNA: Binding of sequence

    specific intercalators

    There is compelling evidence that cellular DNA is the primary

    target for these drugs. Quantitative microspectrofluorometry

    has been used to show that adriamycin rapidly accumulates

    in the nuclei of living cells and is nearly completely bound to

    DNA.(15) Flow cytometry has shown that daunomycin is

    intercalated into DNA within the cell.(16) Furthermore, the

    surface-enhanced Raman spectrum of adriamycin within

    the nuclei of erythroleukemia cancer cells was found to

    resemble that observed in vitro forthe drug DNA complex.(17)

    Together with many other reports (see below) all this clearly

    suggests that anthracycline antitumor antibiotics are strongly

    associated with nuclear DNA in vivo. From a mechanistic

    point of view, drugs that interact with DNA can be divided into

    50 BioEssays 27.1 BioEssays 27:5056, 2004 Wiley Periodicals, Inc.

    1Institute of Biochemistry and Biophysics, University of Tehran, Iran.2Department of Biochemistry and Microbiology, University of Victoria,

    Canada.

    Funding agency: This work was supported by a grant of the Research

    Council of the University of Tehran to A.R. and by a Canadian

    Institutes of Health Research (CIHR) grant to J.A. (MOP-57718).

    *Correspondence to: Juan Ausio, Department of Biochemistry and

    Microbiology, University of Victoria, PO Box 3055, Petch Building

    Room 220, Victoria, BC, V8W 3P6, Canada. E-mail: [email protected]

    DOI 10.1002/bies.20160

    Published online in Wiley InterScience (www.interscience.wiley.com).

    Review articles

  • 5/23/2018 20160_ftp

    2/7

    two broad categories: intercalating drugs and those that

    chemically bind to DNA. Anthracycline antibiotics are classi-

    fied in the first group. Intercalators bind to DNA by inserting

    their planar chromophores between DNAbases. In addition to

    stacking and hydrophobic interactions, the intercalation can

    be further stabilized by hydrogen bond formation in-between

    the bases of DNA, a situation where the sugar moieties are

    interacting withthe central chromophores of the drug.(9,18)The

    anthraquinone ring intercalates between DNA base pairs, with

    itslongaxis nearly perpendicular to theaxisof thedoublehelix.

    One of the rings acts as an anchor and stabilizes the complex

    through hydrogen bond interactions as the daunosamine

    sugar lies in the minor groove.(18) The occurrence of a single

    positive charge on daunomycin contributes electrostatically to

    the binding.(9)

    Binding of daunomycin and adriamycin to DNA has been

    extensively characterized.(1921) Equilibrium dialysis, ab-

    sorption and fluorescence spectroscopy and phase partition

    methods have shown the occurrence of a negative coopera-

    tive binding isotherm pattern in the formation of the daunomy-cinDNA (or adriamycinDNA) complex.(19,20) However, our

    results and those of others using the same technique have

    demonstrated the occurrence of positive cooperative binding

    isotherms.(2224) These controversial results could be due to

    differences in the DNA sequence and/or experimental condi-

    tions used. The magnitude of the binding constant of

    daunomycin to DNA is strongly dependent on the base pair

    composition of DNA. Both drugs exhibit a strong structural

    specificity in their interaction with DNA. Using equilibrium

    dialysis and DNaseI footprinting techniques Chaires et al.(25,26)

    haveshown thatthe preferredsite of intercalationmust contain

    adjacent GC base pairs and the optimal daunomycin-binding

    site may span over a triplet sequence, constituting of anAT base pair at the 50-position, flanked by two contiguous GC

    base pairs. Physically the drug covers three base pairs

    preferentially 50-A/TCG.(25,26) The preference is due to the

    specificity of the hydrogen-binding interactions. The C-9

    hydroxyl group of daunomycin forms a particularly strong

    double hydrogen bond with N-2 and N-3 of the guanine at the

    center of the preferred triplet-binding site.(27) The sequence

    specificity of the daunomycin DNA interaction(26) also reveals

    that the affinity of daunomycin toward natural DNA increases

    with its GC content. The specificity of daunomycin DNA

    interaction has also been determined by high-resolution

    transcription assays.(9,28,29)

    The kinetic analysis of the formation of the daunomycin

    DNA complex shows that this is a slow and complex

    process.(30,31) The average lifetime of the-binding is approxi-

    mately one second and several steps are required to describe

    the time course of its DNA-binding reaction in contrast to what

    is observed with other intercalators. This complicated binding

    kinetics exhibited by daunomycin is most likely the result of its

    preferential binding to certain DNA sequences.

    Chen et al.(32) have performed computational analysis to

    model the interaction of daunomycin with six duplex hexanu-

    cleotides of defined sequence. They inferred from their calcu-

    lations that the most energetically favorable binding sites for

    these antibiotics were the triplet sequences of 50-TCG and 50-

    ACG.(25,32) However, Chaires et al.(25) have shown that the

    requirement for A or T at the 50 end is not absolute and that

    three contiguous GC base pairs form an acceptable binding

    site although possibly with weaker binding affinity.

    The interaction of daunomycin with left-handed Z-DNA

    has also been the subject of many studies(3335) that have

    confirmeda strong preference forthe B-form of DNA.Allosteric

    conversion from Z-DNA to a right-handed conformation by

    daunomycin has also been reported.(35) Binding of daunomy-

    cin to poly (dGdC) under ionic strength conditions that would

    favor the left-handed Z conformation of this DNA, effectively

    inhibits the B-to-Z transition and reverts Z-DNA conformation

    to theright-handedform.Conversion ofDNA toa form different

    from the standard right-handed B conformation results in a

    dramatic reduced affinity for daunomycin.(35)

    Binding of daunomycin and adriamycin to DNA increases

    Tm of DNA melting point by 308C as saturation of the potential

    binding sites is reached.(19) Increase in Tm values of DNA is

    due to stabilization of DNA double helix resulting from the

    intercalation of the drug and can be used to explain DNA and

    RNA synthesis inhibition, as well as the effects of these drugs.

    Interaction with the chromatin:

    unfolding transitions that lead to aggregation

    It is obvious thata simple intercalationmodel for the interaction

    of the drugs with DNA is not enough to explain their overall

    mechanism of action in the cell. In the cell nucleus, DNA does

    notexistas a nakedstructure butis associated with a variety ofproteins making a nucleoprotein complex called chromatin.(36)

    The mechanism of interaction and possible intracellular

    targets of antitumor drugs are often inferred from studies of

    the affinity of these drugs for free DNA in solution. Yet the

    tertiary structure and environment of DNA in the cell can

    significantly modulate these interactions.

    Equilibrium dialysis, hydrodynamic and electric dichroism

    techniques have been used to investigate the effects of

    anthracycline antibiotics on chromatin. Binding isotherms

    obtained from the interaction of daunomycin with 175 bp

    nucleosomes (depleted of histone H1) at several ionic strength

    conditions suggest a negative cooperative pattern and a

    strongly reduced binding relative to free DNA.(3740) It was

    shown that the affinity of daunomycin for different nucleohis-

    tone complexes is in the following order: DNA>175bp H1/H5

    depleted nucleosomes>nucleosome core particles.(14,38)

    These results are in agreement with those observed for the

    interaction of daunomycin with chromatin containing either

    the full native linker histones (H1 and H5) or depleted of these

    histones.(21) Addition of daunomycin to 175 bp H1-depleted

    Review articles

    BioEssays 27.1 51

  • 5/23/2018 20160_ftp

    3/7

    nucleosomes or 146 bp nucleosome core particles stabilizes

    their low melting transition temperature, which corresponds to

    the regions of the nucleosomal DNA less tightly bound to

    the histone octamer at the flanking ends of the particle. (38)

    Comparative studies between H1-depleted 175 bp nucleo-

    somes and their constitutive DNA using circular dichroism(40)

    and hydrodynamic studies(38) have provided additional useful

    structural information. Binding of anthracyclines increases the

    ellipticity of thepositive band at 275nm in nucleosomesand to

    lesser extent in DNA. While with DNA this increase can be

    attributed to changes in the winding angle of the double helix

    resulting from the intercalation of the drug,(41) the relatively

    larger change observed with nucleosomes is most likely the

    result of DNA additionally becoming freed of the histone

    interaction constraints.(42) Consistent with the changes in

    thermal stability of the nucleosome just described, the region

    of DNA affected should correspond to the entry and exit sites

    into the nucleosome becoming stiffer as a result of the

    intercalation of the anthracycline antibiotic. Such release of the

    DNA flanking ends would result in an increase of the frictionalparameters of the nucleosome as it has been experimentally

    shown by the decrease observed in the sedimentation

    coefficient that accompanies the initial titration of the nucleo-

    somes with the drug. Titration beyond 0.15 molecules of drug/

    bp of DNAresults in a reversal of thesedimentation coefficient

    trends, in fact, it starts to increase beyond this point as a result

    of nucleosome aggregation.(38) The hydrodynamic results

    obtained with native or linker histone-depleted chromatin

    fibers(21) mirror these results and provide a further insight into

    the molecular mechanisms involved in the interaction of

    anthracyclines with chromatin (see Fig. 1).

    The hydrodynamic behavior of chromatin complexes

    has been extensively characterized.(43) The ionic-strength-dependent folding of the polynucleosome fiber is highly

    dominated by the occurrence of linker histones (histone

    H1)(44) and only a limited extent of folding is reached in its

    absence.(43) Upon addition of daunomycin to nativeor to linker

    histone-depleted chromatin complexes, a biphasic transition

    in the sedimentation coefficient is observed in both in-

    stances,(21) similar to what is observed for the individual

    nucleosomes.(38) The inflection point of the transition de-

    creases with the ionic strength and with the presence of linker

    histones. This reflects the decrease of free linker DNA

    available for the interaction with the drug under these con-

    ditions and the preference of anthracycline drugs for free

    versus nucleosomal DNA.(38) Our interpretation of these

    results is shown in the model proposed in Fig. 1B and 1C.

    Intercalation of the drug takes place at the free DNA in the

    linker DNA regions, changing the linker twist(45) and gradually

    extending toward the histone octamer, hence, resulting in an

    unfolded chromatin conformation. Binding of the drug to the

    nucleosomal DNA is impeded by the core histone tails.(46)

    Once a critical amount of DNA has been unraveled, the

    nucleosome subunits startlosing their stabilityand, as a result,

    the nucleoprotein complexes start aggregating in a process

    that is reminiscent of the nucleosome aggregation observed

    in vitro when nucleosomes are trimmed with micrococcal

    nuclease beyond 140 bp.

    The chromatin unfolding transitions and final aggregation

    appears to occur without any noticeable dissociation of

    histones(21) and the conformation of the histone octamer is

    little affected by the whole process as reflected by the lack of

    spectral changes in the far UV region of the CD spectrum. (41)

    Our results also indicate that daunomycin has a destabiliz-

    ing rather than an inhibitory effect on chromatin folding(21) and

    easily explain the unwinding/aggregation process described

    by Cera and Palumbo.(47)

    Binding of anthracyclines to

    chromosomal proteins

    A questionstillto be consideredis: do antitumor drugs alsobind

    to any of theprotein components of chromatin? Unfortunately,

    only a few limited reports have been published besides thosedealing with the interaction of these drugs with topoisome-

    rases.(48) Some recent studies have focused their attentionon

    the interaction of antitumor drugs adriamycin and daunomycin

    with histone H1 (linker histones).

    Using a combination of absorption and fluorescence

    spectroscopy, thermal denaturation and equilibrium dialysis

    techniques,(49,50) it has been shown that similar to what is

    observed for the interactions with DNA,(19) the binding of

    daunomycin to histone H1 stabilizes the protein against

    thermal denaturation.(49) This is in contrast to the binding of

    adriamycin to HMGB1, which destabilizes the protein against

    thermal melting(A Rabbani,RM Finn andJ Ausio, unpublished

    data). The binding isotherms for H1-daunomycin complexalways exhibit a positive cooperativity and point to the

    existence of two binding sites. The occurrence of a negative

    Gibbs free energy suggests that the interaction process is

    spontaneous (exergonic).(50) These results come in support

    of the notion that the protein components of chromatin (i.e.

    histone H1) can also be a target for the activity of these

    antitumor drugs. Indeed, recent results from our own labo-

    ratory have shown that daunomycin is able to specifically

    interact withchromatin-bound histone H1(ARabbani,RM Finn

    and J Ausio, unpublished data). Apart from histone H1, the

    interaction of anthracycline antibiotics withtopoisomerases(48)

    and nonhistone proteins have also been reported.(5153)

    Binding of daunomycin to nuclear nonhistone proteins

    within regulatory regions of the genome (for instance in the

    case of topoisomerases) can interfere with the synthesis of

    macromolecules essential for cell proliferation.(54) Therefore,

    daunomycin may exert a dual effect on cell proliferation both

    regulating and modifying the template activity of DNA that

    would be required for the normal RNA synthesis and DNA

    repair. Mechanistically, such drugs may block the activity of

    Review articles

    52 BioEssays 27.1

  • 5/23/2018 20160_ftp

    4/7

    processive enzyme complexes and/or interfere withthe access

    of transactivating factors to DNA.(55,56) Indeed anthracyclines

    have been shown to inhibit theactivity of topoisomerasesI and

    II(48,57) impairing the DNA repair and DNA replication activities

    of the cell.

    Concerted drug-induced structural changes of

    chromatin leading to apoptosis

    Althoughmost of theinteraction of the anthracyclineantibiotics

    involves intercalation between the bases in DNA, these drugs

    can also alter the structure and function of this molecule (and

    hence chromatin) in many different ways. These structural

    changes may ultimately contribute to the apoptotic process

    induced by these drugs in cancer cells(58,59) (see Fig. 2).

    One such change involves the covalent modification of

    DNA by daunomycin.(60) The formation of anthracyclineDNA

    adducts hasbeenobservedto occur both in vitro andin vivo(60)

    through a mechanism that involves the iron-complex of the

    drug.(61) The formaldehyde production resulting from the

    oxidative stress induced by the drug metal complex(61,62)

    produces a covalent attachment to theG-basesof DNA(61) that

    functions as a virtual interstrand crosslinker.(62) Such DNA

    crosslinking could participate in the chromatin aggregation

    process occurring during apoptosis.

    Figure 1. A: Hydrodynamic analysis of the changes in chromatin conformation. The sedimentation coefficient (s20,w) of chromatin (*),

    linker histone-depleted chromatin (*) and corresponding DNA (~) are plotted as a function of the daunomycin:bp ratio (r). The analysis

    shown was performed at75 mMNaCl in10 mMTrisHCl (pH 7.5), 0.1 mMEDTA buffer. B: As daunomycin binds (intercalates to DNA) in

    thelinkerregionsof chromatin,it alters thelinkertwist (redarrows)(45) changingthe orientationof adjacentnucleosomes. C: (I) Chromatin is

    in a folded nativelike conformationunder the experimental ionicconditions usedin A.(43) Thisstructure correlates withthe portionsof the

    plots illustratedin A) andlabeledas 1. (II) The decreaseobserved in the sedimentation coefficient of chromatin as r increases, is indicative

    of a chromatin unfolding. This unfolded chromatin correlates with the portions of the plots illustrated in A) that are labeled as 2. This is the

    result fromthe daunomycin(shownin red)intercalationin thelinkerDNA (seeFig. 1B).(III) Furtherincrease in r resultsin a sudden increase

    in s20,w[see the portions of the plots illustrated in A) that are labeled 3] which is due to aggregation of the chromatin complexes.

    Review articles

    BioEssays 27.1 53

  • 5/23/2018 20160_ftp

    5/7

    The chelation of ions by anthracyclines can also result in

    the generation of reactive oxygen species (ROS), which is

    responsible for the free radical cytotoxicity of these drugs.(63)

    As it occurs with other quinines, these antibiotics can be

    enzymatically reduced to semiquinone radicals, which, in the

    presence of oxygen and iron, can result in the production of

    highly DNA damaging OH-radicals.(63)

    The intercalation process itself results in a distortion of

    the DNA conformation that causes the inhibition of Topo II.(64)

    The effects of anthracyclines on this enzyme(65) have been

    extensively studied (see(66) for a review). In this instance, the

    mechanism of action involves the binding of the drug to the

    Topo IIaDNA complexes forminga ternary complex.(67)Such

    binding interferes with the re-ligation step of the topoisome-

    rase resulting in double-stranded DNA breaks.(66)

    Concluding remarks

    As has been described in the previous sections, DNA

    intercalation of anthracyclines leads to changes in chromatin

    supercoiling that eventually result in chromatin aggrega-

    tion.(23) DNA intercalation still remains one of the main

    targeting sites for the binding of these drugs to DNA.

    Therefore, in closing, we would like to propose that such

    aggregation, in addition to the chromatin structural changes

    described above, may contribute in a concerted way to the

    apoptotic cell death induced by these drugs.(58,59) Chromatin

    aggregation(23) may facilitate or promote the DNA fragmenta-

    tion(68,69) by specific apoptotic endonucleases. Indeed, it has

    been recently shown that chromatin aggregation precedes

    oligonucleosomal fractionation(70) andthatDNA is supercoiled

    in apoptotic chromatin.(71)

    Despite all this, the reason(s) why or how anthracyclines

    exerttheir preferential killing of tumor versus non-carcinogenic

    cells are still not well understood. It is likely that the manifold

    effects described above not only mediate the action of these

    drugs in killing cancer cells but are also responsible for their

    inherent side effects e (i.e. cardiotoxicity).(4,5) A line of thought

    is that their selectivity for the cancer cell targeting resides in

    Figure 2. Anthracycline DNA interaction results in manifold alterations of chromatin structure. The concerted action of these changes

    are most likely directly or indirectly responsible for the chromatin aggregation that precedes the chromatin fragmentation that is

    characteristic of apoptotic cells. The red arrow simply points to intercalation being one of the major components of the anthracycline drug

    DNA interactions.

    Review articles

    54 BioEssays 27.1

  • 5/23/2018 20160_ftp

    6/7

    the lower ability of the cancer cells to repair the damage

    introduced by the drugs, which ultimately results in cell

    apoptosis. New formulations are being currently used to

    enhance the tumor selectivity of anthracyclines using lipo-

    some-cotaining complexes.(72,73) The effort currently devoted

    to the development of these drugs underscores their en-

    ormous potential as anti-tumor agents.

    References1. Arcamone F. 1981. Doxorubicin, anticancer antibiotic. New York:

    Academic Press.

    2. Behal V. 2000. Bioactive products from Streptomyces. Adv Appl

    Microbiol 47:113156.

    3. Myers CE, Mimnaugh EG, Yeh GC, Sinha BK. 1988. Biochemical

    mechanisms of tumor cell kill by the anthracyclines. In: Lown J.W, editor.

    Anthracycline and Anthracenedione-based anticancer agents. pp 527

    569, Amsterdam: Elsevier.

    4. Gianni L, Cordon BJ, Myers CE. 1983. The biochemical bases of

    anthracycline toxicityand antitumor action. In: Rev. BiochemToxicol Vol5.

    Hodgson E, BendJR, Philipot RM,editors. Amsterdam:Elsevier. pp 182.

    5. Umov FD. 2003. Chromatin as a tool for study of genome functions in

    cancer. Ann NY Acad Sci 983:521.

    6. Taatjes DJ, Fenick DJ, Koch TH. 1998. Epidoxoform: a hydrolytically

    more stable anthracycline-formaldehyde conjugate toxic to resistant

    tumor cells. J Med Chem 41:13061314.

    7. Hartmann JT. 2002. Long-term effects of platin and anthracycline

    derivatives and possible prevention strategies. Front Radiat Ther Oncol

    37:92100.

    8. Borchmann P, Morschhauser F, Parry A, Schnell R, Harousseau JL, et al.

    2003. Phase-II study of the new aza-anthracenedione, BBR 2778, in

    patients with relapsed aggressive non-Hodgkins lymphomas. Haema-

    tologica 88:888894.

    9. Chaires JB. 1996. Molecular recognition of DNA, in: Advances in DNA

    Sequence-specific agents, Hurley LH, Chaires JB. editors. Greenwich,

    CT: JAI press. Vol 2. pp 141167

    10. Buchner T, Berdel WE, Hiddemann W. 2004. Priming with granulocyte

    colony-stimulating factorrelation to high-dose cytarabine in acute

    myeloid leukemia. N Engl J Med 350:22152216.

    11. Carrion C, de Madariaga MA, Domingo JC. 2004. In vitro cytotoxic study

    of immunoliposomal doxorubicin targeted to human CD34() leukemic

    cells. Life Sci 75:313328.

    12. Studzian K, Wasowska M, Piestrzeniewicz MK, Wilmanska D, Szmigiero L,

    et al. 2001. Inhibition of RNA synthesis in vitro and cell growth by

    anthracycline antibiotics. Neoplasma 48:412 418.

    13. Leng F, Leno GH. 1997. Daunomycin disrupts nuclear assembly and the

    coordinate initiation of DNA replication in Xenopus egg extracts. J Cell

    Biochem 64:476491.

    14. Fritzsohe H, Wahnert V, Chaires JB, Dattagupta N, Schlessinger FB, et al.

    1987. Anthracycline antibiotics interaction with DNA and nucleosomes

    and inhibition of DNA synthesis. Biochemistry 26:19962000.

    15. Gigli M, Doglia SM, Millet JM, Valentini L, Manfait M. 1988. Quantitative

    study of doxorubicin in living cell nuclei by microspectrofluorometry.

    Biochem Biophys Acta 950:1320.

    16. Belloc F, Lacombe F, Dumain P, Lopez F, Bernard P, et al. 1992.

    Intercalation of anthracyclines into living cell DNA analyzed by flow

    cytometry. Cytometry 13:880 885.

    17. Nabiev I, Sokolov K, Morjani H, Manfait M. 1991. Surface-enhanced

    Raman spectroscopy of nucleic acids and antitumour drug/nucleic acid

    complexes in vitro and in living cells. In: Hester RE, Griling RB, editor.

    Spectroscopy of biological molecules. York, UK: The Royal Society of

    Chemistry. pp 345348.

    18. Wang AH-J. 1993. Structure-activity studies of anthracyclineDNA com-

    plexes. In: Neidle S, Waring M, editors. Molecular Aspects of Anticancer

    Drug-DNA Interactions. Vol 1. Boca Raton, FL: CRC Press. pp 3253.

    19. Chaires JB, Dattagupta ND, Crothers DM. 1982. Studies on interaction of

    anthracycline antibiotics and DNA: equilibrium binding studies on

    interaction of daunomycin with DNA. Biochemistry 21:39333940.

    20. BarceloF, Martorell J, Gavilanes F, Gonzalez-Ros JM. 1988. Equilibrium

    binding of daunomycin and adriamycin to calf thymus DNA. Tempera-

    ture and ionic strength dependence of thermodynamic parameters.

    Biochem Pharmacol 37:21332138.

    21. Chaires JB, Satyanarayana S, Suh D, Fokt I, Przewloka T, Priebe W.

    1996. Parsing the free energy of anthracycline antibiotic binding to DNA.

    Biochemistry 35:20472053.

    22. Graves DE, Krugh TR. 1983. Adriamycin and daunomycin bind in a

    cooperative manner. Biochemistry 22:3941 3947.

    23. Rabbani A, Iskandar M, AusioJ. 1999. Daunomycin induced unfolding

    and aggregation of chromatin. J Biol Chem 274:1840118406.

    24. Rabbani A, Hagsharifi-Taghavi M, Goliaei B. 1993. Binding of the anti-

    tumor drug adriamycin to DNA-histone complexes. Med J IRI 6:275 279.

    25. Chaires JB, Herrera JE, Waring MJ. 1990. Preferential binding of

    daunomycin to 50ATCG and 50ATGC sequences revealed by footprint-

    ing, titration experiment. Biochemistry 29:61456153.

    26. Chaires JB, Fox KR, Herrera JE, Britt M, Waring MJ. 1987. Site and

    sequence specificity of the daunomycin-DNA interaction. Biochemistry

    26:82278236.

    27. Chen KX, Gresh N, Pullman B. 1986. A theoretical investigation on the

    sequence selected binding of adriamycin to double stranded poly-

    nucleotide. Nucleic Acid Res 14:22512267.

    28. Chaires JB. 1990. Daunomycin binding to DNA: from the macroscopic to

    the microscopic. In: Pullman B, Jortner J, editor. Molecular basis of

    specificity in nucleic acid-drug interaction. Dordrecht, the Netherlands:

    Kluwer Academic Press. pp 123136.

    29. Skorobogaty A, White RJ, Phillips DR, Reiss JA. 1988. Elucidation of the

    DNA sequence preferences of daunomycin. Drug Design and Delivery

    3:125152.

    30. Rizzo V, Sacchi N, Menozzi M. 1989. Kinetic studies of anthracycline-

    DNA interaction by fluorescence stopped flow confirm a complex

    association mechanism. Biochemistry 28:274282.

    31. Philips DR, Greif PC, Boston RC. 1988. Daunomycin-DNA dissociation

    kinetics. Mol Pharmacol 33:225230.

    32. Chen KX, Cresh N, Pullman B. 1985. A theoretical investigation on the

    sequence selective binding of daunomycin to double stranded poly-

    nucleotide. J Biomol Struct Dyn 3:445466.

    33. Britt M, Zunine F, Chaires JB. 1986. The interaction of the beta-anomer of

    doxorubicin with B and Z DNA. Mol Pharmacol 29:7480.

    34. Chaires JB. 1983. Danomycin inhibits the B leads to Z transition in poly

    d(G-C). Nucleic Acids Res 11:84858494.

    35. Chaires JB. 1986. Allosteric conversion of Z-DNA to an intercalated right-

    handed conformation by Daunomycin. J Biol Chem 261:88998907.

    36. van Holde KE. 1988. Chromatin, New York: Springer-Verlag.

    37. Simpkins H, Pearman LF, Thompson LM. 1984. Effect of adriamycin on

    supercoiled DNA and nucleosomes. Cancer Res 44:613618.

    38. Chaires JB, Dattagupta N, Crothers DM. 1983. Binding of daunomycin to

    calf thymus nucleosomes. Biochemistry 22:284292.

    39. Portugal J, Waring MJ. 1987. Analysis of the effects of antibiotics on the

    structure of nucleosome core particle determined by DNase I cleavage.

    Biochemie 69:825840.

    40. Cera C, Palu G, Magno SM, Palumbo M. 1991. Interaction between

    second generation anthracyclines and DNA in the nucleosome structure.

    Nucleic Acids Res 19:23022314.

    41. Johnson BP, Dahl KS, Tinoco I, Ivanov V, Zurkin VB. 1981. Correlations

    between deoxyribonucleic acid structural parameters and calculated

    circular dichroism spectra. Biochemistry 20:7379.

    42. Greulich KO, Ausio J, Eisenberg H. 1985. Nucleosome core particle

    structure and structural changes in solution. J Mol Biol 186:167173.

    43. Ausio J. 2000. Analytical ultracentrifugation and characterization of

    chromatin structure. Biophysical Chem 86:141153.

    44. Ramakrishnan V. 1997. Histone H1 and chromatin higher-order structure.

    Crit Rev Eukaryot Gene Expr 7:215230.

    45. Krajewski WA, Ausio J. 1996. Modulation of the higher-order folding of

    chromatin by deletion of histone H3 and H4 terminal domains. Biochem J

    316:395400.

    46. Mir MA, Das S, Dasgupta D. 2004. N-terminal tail domains of core

    histones in nucleosome block the access of anticancer drugs,

    mithramycin and daunomycin, to the nucleosomal DNA. Biophys Chem

    109:121135.

    Review articles

    BioEssays 27.1 55

  • 5/23/2018 20160_ftp

    7/7

    47. Cera C, Palumbo M. 1990. Anticancer activity of anthracycline antibiotics

    and DNA condensation. Anti-cancer drug design 5:265271.

    48. Capranico G, Zunino F, Kohn KW, Pemmier Y. 1990. Sequence-selective

    topoisomerase II inhibition by anthracycline derivatives in SV40 DNA:

    relationship with DNA binding affinity and cytotoxicity. Biochemistry

    29:562569.

    49. Zargar SJ, Rabbani A. 2000. The effects of daunomycin antibiotic on

    histone H1: thermal denaturation and fluorescence spectroscopy

    studies. Int J Biol Macromol 28:7579.

    50. Zargar SJ, Rabbani A. 2002. Interaction of daunomycin antibiotic with

    histone H1: ultraviolet spectroscopy and equilibrium dialysis studies. Int

    J Biol Macromol 30:113117.

    51. Rabbani A, Sharoghi B, Khodabandeh M. 1995. Interaction of doxor-

    ubicin with DNA-HMG1 complex. J Sci IRI 6:147152.

    52. Kikuchi H, Sato S. 1978. Fractionation of daunomycin to nonhistone

    proteins from rat liver. Biochem Biophys Acta 532:113121.

    53. Kikuchi H, Sato S. 1978. Binding of daunomycin to nonhistone proteins

    from rat liver. Biochem Biophys Acta 434:509512.

    54. Campiglio M, Somenzi G, Olgiati C, Beretta G, Balsari A, et al. 2003. Role

    of proliferation in HER2 status predicted response to doxorubicin. Int J

    Cancer 104:568573.

    55. Gniazdowski M, Denny WA, Nelson SM, Czyz M. 2003. Transcription

    factorsas targetsfor DNA-interacting drugs. Curr MedChem10:909924.

    56. Peterson M, Tupy J. 1994. Transcription factors: a new frontier in

    pharmaceutical development. Biochem Pharmacol 47:127 128.

    57. Lothstein L, Israel M, Sweatman TW. 2001. Anthracycline drug targeting:

    cytoplasmic versus nucleara fork in the road. Drug Resist Updat 4:

    169177.

    58. Lee TK, Lau TC, Ng IO. 2002. Doxorubicin-induced apoptosis and

    chemosencitivity in hepatoma cell lines. Cancer Chemother Pharmacol

    49:7886.

    59. Hale AJ, Smith CA, Sutherland LC. 1996. Apoptosis: molecular

    regulation of cell death. Eur J Biochem 236:126.

    60. Purewal M, Liehr JG. 1993. Covalent modification of DNA by daunor-

    ubicin. Cancer Chemother Pharmacol 33:239244.

    61. Taatjes DJ, Koch TH. 2001. Nuclear targeting and retention of anthracy-

    cline antitumor drugs in sensitive and resistant tumor cells. Curr Med

    Chem 8:1529.

    62. Parker BS, Cullinane C, Phillips DR. 1999. Formation of DNA adducts

    by formaldehyde-activated mitoxantrone. Nucleic Acids Res 27:2918

    2923.

    63. Muller I, Niethammer D, Bruchelt G. 1998. Anthracycline-derived

    chemotherapeutics in apoptosis and free radical cytotoxicity. Int J Mol

    Med 1:491494.

    64. Tewey KM, Rowe TC, Yang L, Halligan BD, Liu LF. 1984. Adriamycin-

    induced DNA damage mediated by mammalian DNA topoisomerase II.

    Science 226:466468.

    65. MacGrogan G, Rudolph P, Mascarel IdI, Mauriac L, Durand M, et al.

    2003. DNA topoisomerase IIalpha expression and the response to

    primary chemotherapy in breast cancer. Br J Cancer 89:666671.

    66. Kellner U, Sehested M, Jensen PB, Gieseler F, Rudolph P. 2002. Culprit

    and victimDNA topoisomerase II. Lancet Oncol 3:235243.

    67. Binaschi M, Bigioni M, Cipollone A, Rossi C, Goso C, et al. 2001.

    Anthracyclines: selected new developments. Curr Med Chem Anti-Canc

    Agents 1:113130.

    68. Levin M, Silber R, Israel M, Goldfeder A, Khetarpal VK, et al. 1981.

    Protein-associated DNA breaks and DNA-protein cross links by DNA

    nonbinding derivatives of adrimycin in L1210 cells. Cancer Res

    41:10061010.

    69. Zwelling LA, Michael S, Erickson LC, Unserleider RS, Nichols M, et al.

    1981. Protein-associated deoxyribonucleic acid strand breaks in L1210

    cells treated with the deoxyribonucleic acid intercalating agents 40-(9-

    acridinylamino) methanesulfon-m-anisidide and adriamycin. Biochemis-

    try 20:65536563.

    70. Hendzel MJ, Nishioka WK, Raymond Y, Allis CD, Bazett-Jones DP, Thng

    JP.1998. Chromatin condensationis notassociatedwith apoptosis. J Biol

    Chem 273:2447024478.

    71. DArrigo C, Barboro P, Mormino M, Coradeghini R, Parodi S, et al. 2003.

    DNA supercoiling in apoptotic chromatin. Biochem Biophys Res Comm

    309:540546.

    72. Hamaguchi T, Matsumura Y, Nakanishi Y, Muro K, Yamada Y, et al. 2004.

    Antitumor effect of MCC-465, pegylated liposomal doxorubicin tagged

    with newly developed monoclonal antibody GAH, in colorectal cancer

    xenografts. Cancer Sci 95:608613.

    73. Wolff AC. 2003. Liposomal anthracyclines and new treatment ap-

    proaches for breast cancer. Oncologist 8(Suppl 2):2530.

    Review articles

    56 BioEssays 27.1