dna damage helle

Upload: divassamp

Post on 03-Jun-2018

229 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/11/2019 Dna Damage Helle

    1/12

    Cancer therapy usually involves exposing the body toagents that kill cancer cells more efficiently than normaltissue cells. Such therapies must therefore exploit specificmolecular and cellular features of the cancer they are aim-ing to eliminate. Most cancer cells proliferate more rapidlythan their normal counterparts so most cancer drugstarget the cell cycle. Cell division can be targeted directlyby inhibitors of the mitotic spindle, thus preventing equaldivision of DNA to the two daughter cells. The growth sig-nals that result in entry into the cell cycle can be targetedby hormonal manipulation, therapeutic antibodies anddrugs that inhibit growth signalling pathways. However,the most common means of targeting the cell cycle is toexploit the effect of DNA-damaging drugs. DNA damagecauses cell-cycle arrest and cell death either directly orfollowing DNA replication during the S phase of the cellcycle. Cellular attempts to replicate damaged DNA cancause increased cell killing, thus making DNA-damagingtreatments more toxic to replicating cells than to non-

    replicating cells. However, the toxicity of DNA-damagingdrugs can be reduced by the activities of several DNArepair pathways that remove lesions before they becometoxic. The efficacy of DNA damage-based cancer therapycan thus be modulated by DNA repair pathways. In addi-tion, some of these pathways are inactivated in some cancertypes. These two features make DNA repair mechanisms apromising target for novel cancer treatments.

    DNA-damaging agents in cancer treatment

    Many cancer drugs employed in the clinic have beenused for several decades and are highly efficient in kill-ing proliferating cells (FIG. 1). High levels of DNA damage

    cause cell-cycle arrest and cell death. Furthermore, DNAlesions that persist into the S phase of the cell cycle canobstruct replication fork progression, resulting in theformation of replication-associated DNA double-strandbreaks (DSBs). DSBs are generally considered to be themost toxic of all DNA lesions1,2.

    Common types of DNA damage that interfere withreplication fork progression are chemical modifications(adducts) of DNA bases, which are created by reactivedrugs that covalently bind DNA either directly or afterbeing metabolized in the body. These alkylating agents aregrouped in two categories: monofunctional alkylatingagents with one active moiety that modifies single basesand bifunctional alkylating agents that have two reactivesites and crosslink DNA with proteins or, alternatively,crosslink two DNA bases within the same DNA strand(intra-strand crosslinks) or on opposite DNA strands(inter-strand crosslinks). Inter-strand crosslinks pose asevere block to replication forks.

    DNA synthesis is sometimes targeted by inhibitorsof DNA replication, such as aphidicolin, which directlyinhibits DNA polymerases3, whereas the radical scav-enger hydroxyureainhibits ribonucleotide reductase,which is required for production of the dNTPs that areused for DNA synthesis4. These two replication inhibi-tors can be regarded as DNA-damaging agents becausethey impair replication fork progression and cause DNAlesions, including DSBs5,6.

    Antimetabolites, such as5-fluorouracil(5FU) and thio-purines, resemble nucleotides, nucleotide precursors orcofactors required for nucleotide biosynthesis and actby inhibiting nucleotide metabolism pathways, thus

    *Radiation Oncology &

    Biology, University of Oxford,Old Road Campus Research

    Building, off Roosevelt Drive,

    Headington, Oxford,

    OX3 7DQ, UK.Department of Genetics

    Microbiology and Toxicology,

    Stockholm University, Svante

    Arrhenius vg 16, S-106 91

    Stockholm, Sweden.

    Correspondence to T.H.

    e-mail: thomas.helleday@rob.

    ox.ac.uk

    doi:10.1038/nrc2342

    Published online

    7 February 2008

    Alkylating agents

    Electrophilic compounds that

    are reactive either directly or

    following metabolism and bind

    covalently to electron-rich

    atoms in DNA bases (that is,

    oxygen and nitrogen).

    Antimetabolites

    Compounds with similar

    chemical structures to

    nucleotide metabolites that

    interfere with nucleotide

    biosynthesis or are

    incorporated into DNA.

    DNA repair pathways as targetsfor cancer therapyThomas Helleday*, Eva Petermann*, Cecilia Lundin*, Ben Hodgson*and

    Ricky A. Sharma*

    Abstract | DNA repair pathways can enable tumour cells to survive DNA damage that is

    induced by chemotherapeutic treatments; therefore, inhibitors of specific DNA repair

    pathways might prove efficacious when used in combination with DNA-damaging

    chemotherapeutic drugs. In addition, alterations in DNA repair pathways that arise duringtumour development can make some cancer cells reliant on a reduced set of DNA repair

    pathways for survival. There is evidence that drugs that inhibit one of these pathways in

    such tumours could prove useful as single-agent therapies, with the potential advantage

    that this approach could be selective for tumour cells and have fewer side effects.

    R E V I E W S

    NATURE REVIEWS |CANCER VOLUME 8 |MARCH 2008 |193

    http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=40685http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=43130http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=43130mailto:[email protected]:[email protected]:[email protected]:[email protected]://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=43130http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=40685
  • 8/11/2019 Dna Damage Helle

    2/12

    Non-homologous end

    joining

    Connection and resealing of

    the two ends of a DNA double-

    strand break without the need

    for sequence homology

    between the ends.

    Homologous recombinationA process that can copy a DNA

    sequence from an intact DNA

    molecule (often the newly

    synthesized sister chromatid)

    to repair or bypass replication

    lesions.

    Base-excision repair

    A repair pathway that replaces

    missing or modified DNA

    bases, such as those produced

    by alkylating agents or in

    spontaneously degraded DNA,

    with the correct DNA base.

    Nucleotide-excision repairA process that removes large

    DNA adducts or base

    modifications that distort the

    double helix and uses the

    opposite strand as template

    for repair.

    Alkyltransferases

    A class of enzymes that

    directly reverse DNA base

    modifications that are induced

    by alkylating agents by

    transferring the alkyl group

    from the base onto the

    protein.

    depleting cells of dNTPs. They can also impair replicationfork progression by becoming incorporated into the DNA7.In general, the molecular mechanisms through whichanti-metabolites induce cell death are poorly understood.

    Another means of interfering with replication is toexploit DNA strand breaks that arise naturally during theprocess of DNA synthesis. Topoisomerases are a groupof enzymes that resolve torsional strains imposed onthe double helix during DNA replication. They inducetransient DNA breaks to relax supercoiled DNA or allowDNA strands to pass through each other8. Resealing ofthese breaks can be prevented by the use of topoisomer-ase poisons that trap the enzymes in complex with theDNA. The nature of the damage that is caused dependson which type of enzyme is targeted. Topoisomerase IIpoisons cause DSBs, and topoisomerase I poisons causepositive supercoils in advance of replication forks134and replication-associated DSBs1,2. This is a strategycommonly used for cancer treatment.

    Ionizing radiation and radiomimetic agents such asbleomycincause replication-independent DSBs that cankill non-replicating cells. In addition, such treatmentscan also rapidly prevent DNA replication by activation of

    cell-cycle checkpoints to avoid formation of toxic DNAreplication lesions9.

    Cell-cycle checkpoints are regulated by effector kinases,such as ataxia telangiectasia mutated (ATM) and ATMand Rad3-related (ATR)1012, which regulate the activitiesof downstream checkpoint proteins, such as checkpointkinases 1 (CHK1) and 2 (CHK2). Defects in DNA dam-age checkpoint pathways result in sensitivity to a range ofanticancer treatments, for example, loss of ATM results insensitivity to ionizing radiation13. The triggering of thesecheckpoints and subsequent DNA repair activity largelydetermines the efficacy of anticancer drugs in causingtumour regression.

    Efficient repair of chemotherapy lesions

    Direct DSBs are mainly repaired by non-homologousend joining14, whereas replication-associated DSBsare repaired by homologous recombination(HR)15andrelated replication repair pathways. DNA adducts, suchas those created by alkylating agents, may be excisedand repaired before they are confronted by the replica-tion machinery. This is achieved by base-excision repair,excising a single damaged DNA base or a short strandcontaining the damaged base16or nucleotide-excisionrepair(NER), which excises a single-stranded DNAmolecule of approximately 2430 base pairs contain-ing the DNA lesion17,18. Damaged DNA can also berepaired without removal of the damaged base, in aprocess that directly reverses the DNA alkylation19. TheO-6-methylguanine-DNA methyltransferase (MGMT)is an alkyltransferase that removes alkylations on the O6position of guanine produced by anticancer drugs suchas temozolomide20, and the DNA dioxygenasesABH2(also known as ALKBH2) and ABH3 (also knownas ALKBH3) revert 1-methyladenine and 3-methyl-

    cytosine back to adenine or cytosine respectively21. Therepair of alkylated lesions is thought to be quick, withthe majority of lesions probably being repaired withinone hour. If the lesions are removed before the initia-tion of replication, the efficiency of alkylating agents inkilling the tumour is significantly reduced. Thus, modu-lation of DNA repair clearly influences the efficacy ofalkylating agents, and resistance to alkylating agents isoften explained by increased expression and/or activityof DNA repair proteins.

    Whereas most DNA repair pathways mediateresistance to DNA damage, mismatch repairis actuallyrequired for the toxicity of several anticancer drugs(FIG. 1). This has been explained by the futile repaircycle model in which mismatch repair removes thenewly inserted intact base instead of the damaged base,triggering subsequent rounds of futile repair which canbe deleterious to the cell22. It is also possible that mis-match repair might have an important role in triggeringcheckpoint signalling and apoptosis, which might medi-ate increased cytotoxicity23. It has been established thata defect in mismatch repair is associated with resistanceto many, but not all, DNA-damaging anticancer agents,such as monofunctional alkylating agents and cispla-tin, as well as the antimetabolite 6-thioguanine7,22,24. Itshould be noted that mismatch repair acts directly atreplication forks and can therefore not prevent them

    from encountering damage.Collapse of replication forks during DNA synthesis

    can be avoided by bypassing DNA lesions in a processcalled translesion synthesis25,26. This process is carried outby switching the regular polymerases, and , whichare responsible for leading and lagging strand synthesis,respectively27,28, to polymerases with different substratespecificities, thus enabling them to bypass different typesof damaged bases29.

    Once a replication fork stalls or collapses, otherrepair pathways are required to permit resumption ofreplication. Collapsed replication forks are recognizedby the checkpoint machinery, which will in turn trigger

    At a glance

    Several cancer chemotherapy drugs work by producing excessive DNA damage that

    causes cell death directly or following DNA replication. Survival is promoted through

    repair of these lesions by a number of DNA repair pathways.

    The efficacy of anticancer drugs is highly influenced by cellular DNA repair capacity.

    Inhibitors of DNA repair increase the efficacy of DNA-damaging anticancer drugs in

    preclinical models. Small-molecule inhibitors of DNA repair have been combined

    with conventional chemotherapy drugs in several phase III clinical trials.

    Tumour development can be associated with perturbed DNA damage response and

    repair pathways. This perturbation results in reduced DNA repair capacity and

    increased genetic instability in tumour cells. Defects in one DNA repair pathway can

    be compensated for by other pathways. Such compensating pathways can be

    identified in synthetic lethality screens and then specifically targeted for treatment

    of DNA repair-defective tumours.

    Evidence indicates that inhibitors of DNA repair pathways can work as single agents

    for the targeted treatment of DNA repair-defective cancers. This hypothesis is

    currently being tested in phase II trials in which patients with breast or ovarian

    cancers that are defective in homologous recombination are being treated with a

    poly(ADP-ribose) polymerase inhibitor.

    Tumours often exhibit replication stress as a consequence of oncogene-induced

    growth signals or hypoxia-induced replication arrest. We propose that DNA repair

    inhibitors could be used to prevent the repair of replication lesions present in tumourcells and convert them into fatal replication lesions that specifically kill cancer cells.

    R E V I E W S

    194 |MARCH 2008 |VOLUME 8 www.nature.com/reviews/cancer

    http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39139http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=472http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=545http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=1111http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=11200http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=4255http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=121642http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=221120http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39515http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39515http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=43669http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=43669http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39515http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39515http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=221120http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=121642http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=4255http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=11200http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=1111http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=545http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=472http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39139
  • 8/11/2019 Dna Damage Helle

    3/12

    |

    OH

    Toxic lesions

    Single-strand breaks

    Double-strand breaks

    Base damage

    Includes mismatchrepair-mediatedtoxicity

    No

    Major repair pathways

    HR

    HR

    HR

    HR

    HR

    YesAlkylsulphonates

    Nitrosourea compounds

    Temozolomide

    b Monofunctional alkylators

    c Bifunctional alkylators

    Nitrogen mustard

    Mitomycin C

    CisplatinNER

    NER

    AT

    Yes

    ?

    Cancer treatment

    Yes BER

    BER

    BER

    d Antimetabolites

    5-Fluorouracil (5FU)

    Thiopurines

    Folate analogues

    NHEJ

    NHEJ

    NHEJ

    SSBR

    SSBR

    e Topoisomerase inhibitors

    Camptothecins

    Etoposide (VP16)

    f Replication inhibitors

    Aphidicolin

    Hydroxyurea

    a Radiotherapy and radiomimetics

    Ionizing radiation

    Bleomycin

    TLS

    TLS

    No

    No

    FA

    FA

    FA

    FA

    O2G

    RecQ

    RecQ

    RecQ

    RecQ

    Double-strand breaks

    Replication lesions

    Double-strand breaks

    Single-strand breaks

    Replication lesions

    Double-strand breaks

    DNA crosslinks

    Bulky adducts

    Replication lesions

    Base damage

    Bulky adducts

    Replication lesions

    ENDO

    ENDO

    ENDO

    ENDO

    Replication lesions

    Uncharacterized

    Base damage

    Pt

    H3CS

    O

    OO

    H3CS

    OCH3

    OO

    CH3

    N

    N

    NH

    N

    SH

    H2N

    H3NPt

    NH3

    Cl Cl

    DNA dioxygenases

    A class of enzymes that

    directly reverse DNA base

    methylations through an

    oxidation mechanism. The

    human DNA dioxygenase

    ABH2 is thought to act at

    replication forks.

    Mismatch repair

    A process that acts during

    DNA replication to correct

    base-pairing errors made by

    the DNA polymerases.

    Figure 1 |Overview of DNA repair pathways involved in repairing toxic DNA lesions formed by cancer

    treatments. The DNA-damaging agents that are used in cancer treatment induce a diverse spectrum of toxic DNA

    lesions. These lesions are recognized by a variety of DNA repair pathways which are lesion-specific but are

    complementary in some respects. a| Ionizing radiation and radiomimetic drugs induce double-strand breaks

    (DSBs) that are predominantly repaired by non-homologous end joining (NHEJ). b, c| Monofunctional alkylators (b)

    and bifunctional alkylators (c) induce DNA base modifications, which interfere with DNA synthesis. Lesionsproduced by some alkylators are processed into toxic lesions in a mismatch repair-dependent manner. The base-

    excision repair (BER) and nucleotide-excision repair (NER) pathways are, together with alkyltransferases (ATs),

    major repair pathways, whereas other repair pathways repair toxic replication lesions, such as those produced by

    interstrand crosslinks. d| Antimetabolites interfere with nucleotide metabolism and DNA synthesis, causing

    replication lesions which have not yet been characterized. Mismatch repair mediates the toxicity of some

    antimetabolites (for example, thiopurines). The repair pathways involved in repair of antimetabolite-induced

    lesions are, apart from BER, poorly characterized. e| Topoisomerase poisons trap topoisomerase I or II in transient

    cleavage complexes with DNA, thus creating DNA breaks and interfering with replication. f| Replication inhibitors

    induce replication fork stalling and collapse, resulting in indirect DSBs. The relative contributions of the major

    repair pathways to the respective types of DNA damage outlined are indicated by the sizes of the boxes. This is

    based on the extent of sensitivity of repair-deficient cells to anticancer drugs in each category. ENDO, endonuclease-

    mediated repair; FA, Fanconi anaemia repair pathway; HR, homologous recombination; O2G, DNA dioxygenases;

    RecQ, RecQ-mediated repair; SSBR, DNA single-strand break repair; TLS, translesion synthesis.

    R E V I E W S

    NATURE REVIEWS |CANCER VOLUME 8 |MARCH 2008 |195

  • 8/11/2019 Dna Damage Helle

    4/12

    Translesion synthesis

    A mechanism during DNA

    replication in which the

    standard DNA polymerase is

    temporarily exchanged for a

    specialized polymerase that

    can synthesize DNA across

    base damage on the template

    strand.

    Fanconi anaemia repair

    pathway

    Proteins of this pathway,

    including BRCA2, are mutated

    in the hereditary disorder

    Fanconi anaemia (FA), resulting

    in hypersensitivity to inter-

    strand crosslinks. Evidence

    suggests that the FA pathway

    promotes the repair of stalled

    replication forks, possibly by

    activating HR and facilitating

    ATR- and ATM-dependent

    checkpoint signalling.

    Endonuclease-mediated

    repair

    A repair pathway that

    introduces a DNA single-strand break in a DNA

    structure to facilitate

    continuous repair.

    RecQ-mediated repair

    A repair pathway that unwinds

    complex DNA structure to

    facilitate repair.

    Therapeutic index

    The therapeutic index

    describes the ability of a

    treatment strategy to kill

    cancer cells in preference to

    cells in normal tissues.

    cell-cycle arrest12, DNA repair30or cell death throughapoptosis or senescence3133. Although we know littleof the nature of replication lesions, there is an increas-ing body of information concerning pathways thatrepair them. HR has a central role in the repair of mostreplication lesions formed by anticancer drugs5,6,15,34.There are several ways by which HR is used to restart

    replication. The sequence identity between two newlysynthesised DNA molecules can be used to restart rep-lication behind the replication block. Also, recombina-tion can be used to bypass DNA lesions in a processcalled template switching35. Other repair pathwaysactive at replication forks involve the Fanconi anaemia(FA) repair pathway36, endonuclease-mediated repair, suchas that mediated by the MUS81endonuclease37, andRecQ-mediated repair, which involves DNA helicasessuch as Bloom syndrome (BLM)38, Werner syndrome(WRN)39,40and other members of the RecQ family ofhelicases41. Several of the proteins in these pathwayshave been found to be directly linked with HR42or theresolution of recombination products such as Holliday

    junctions38,40,43,44. However, cells that are defective inthese pathways show distinct differences from HR-defective cells, indicating that they represent differentbut overlapping repair pathways45.

    Cells defective in a specific DNA repair pathwayexhibit sensitivity to drugs producing DNA lesions thatare normally repaired by that pathway. This sensitivityhas been exploited to isolate hamster cell lines showinghypersensitivity to cancer treatments such as etoposide,mitomycinCand ionizing radiation, and also to allowcloning of genes involved in DNA repair46. The DNArepair pathways involved in the repair of damage causedby various anticancer agents are summarized in FIG. 1.

    These DNA repair pathways can have increased activityin tumour cells, resulting in resistance to chemotherapeu-tic drugs47. Importantly, these DNA repair pathways canbe inhibited pharmacologically to potentially increase theefficacy or specificity of anticancer agents.

    DNA repair inhibitors in combination therapy

    The basic understanding of DNA repair, from theprinciples of the DNA lesions created to the pathwaysthat are capable of repairing these lesions, has increasedconsiderably during recent years. This knowledgepermits rational combination of cytotoxic agents andinhibitors of DNA repair to enhance tumour-cell killing.

    Understanding lesions and repair pathways enables theuse of DNA-repair inhibitors to exploit tumour defectsor cancer-specific replication lesions (BOX 1). Severalinhibitors of DNA repair have been developed as clinicalagents and clinical trials are ongoing (TABLE 1).

    Sensitizers to alkylating agents. Despite the adverse sideeffects caused by alkylating agents on bone marrow andother normal tissues, drugs such ascyclophosphamide,ifosfamide, chlorambucil, melphalanand dacarbazineremain some of the most commonly prescribedchemotherapies in adults and children with varioussolid and haematological malignancies, particularly incombination with anthracyclines and steroids in multi-agent regimens. More recently, a DNA alkylator andmethylator developed in the 1980s, temozolomide(anoral prodrug that crosses the bloodbrain barrier), haschanged clinical practice in the treatment of high-gradegliomas in adults and children48,49.

    A class of agents currently being tested in clinicaltrials in combination with temozolomide therapy con-

    sists of the pseudosubstrates for MGMT. The lead com-pounds in this class have been O6-benzylguanine50andlomeguatrib (AstraZeneca, London, UK); the latter isalso known as O6-(4-bromothenyl)guanine or PaTrin-2.Resistance to O6-alkylating agents can be overcome inpreclinical models by depletion of MGMT51and a rela-tionship exists between MGMT activity and resistanceto chloroethylating nitrosoureas and methylating agentsin tumour cells grown in vitro and in xenograft mod-els52. O6-Benzylguanine and lomeguatrib have recentlybeen tested in phase III clinical trials and biologicallyeffective doses have been established for both agents53.In the case of O6-benzylguanine, a phase I clinical trialnot only defined the maximum tolerated dose (MTD)of a single dose of temozolomide when combined withO6-benzylguanine, but it also determined the doseof O6-benzylguanine that was effective in producingcomplete depletion of tumour MGMT activity for48 h54. However, results obtained so far indicate that,when used in combination with cytotoxic chemo-therapy, myelosuppression is significantly enhancedby O6-benzylguanine and lomeguatrib, necessitatingsignificant reductions in the doses of alkylating agentsprescribed from those used in standard chemotherapy55.On account of this lack of selectivity for malignant tis-sue versus normal bone marrow, no improvement in thetherapeutic indexhas so far been demonstrated in clinical

    trials of these agents.The combination of temozolomide with inhibitors

    of poly(ADP-ribose) polymerase 1 (PARP1) is currentlyunder investigation in several clinical trials (TABLE 1).PARP1 is required for the efficient base-excision repairof apurinic sites, the intermediate DNA lesions inducedby temozolomide, and inhibition of PARP1 retards therepair of these lesions. It was originally shown in 1980by Durkacz and Shall that specific inhibitors of PARPcan prevent the rejoining of DNA strand breaks that arecaused by dimethyl sulphate, resulting in demonstrablecytotoxicity in vitro56. However, in the absence of PARP1inhibition, apurinic sites are not generally regarded

    Box 1 | Strategies using DNA repair inhibitors in cancer treatment

    DNA repair inhibitors can be used in combination with a DNA-damaging anticancer

    agent. This will increase the efficiency of the cancer treatment by inhibiting DNA

    repair-mediated removal of toxic DNA lesions.

    DNA repair inhibitors can be used as monotherapy to selectively kill cancer cells

    with a defect in the DNA damage response or DNA repair. Synthetic lethal

    interactions between a tumour defect and DNA repair pathway can be used to

    identify novel treatment strategies.

    In the future, DNA repair inhibitors could potentially be used to amplify tumour-

    specific replication lesions to selectively kill cancer cells. This strategy would take

    advantage of cancer-specific replication stress caused by oncogenes or the tumour

    microenvironment.

    R E V I E W S

    196 |MARCH 2008 |VOLUME 8 www.nature.com/reviews/cancer

    http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=80198http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=641http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7486http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39207http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42674http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42674http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42674http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39748http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39748http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=40024http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=43113http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42973http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39768http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=41671http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=142http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=142http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=41671http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39768http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42973http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=43113http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=40024http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39748http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42674http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39207http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7486http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=641http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=80198
  • 8/11/2019 Dna Damage Helle

    5/12

    as major contributors to the cytotoxicity induced bytemozolomide. The success of the treatment rationaleadopted by current clinical trials of GPI-21016 (GuilfordPharmaceuticals, Baltimore, Maryland, USA), INO-1001(Inotek Pharmaceuticals, Beverly, Massachusetts, USA)and AG-014699 (Pfizer GRD, La Jolla, California, USA)depends on the overall biological role of and necessityfor PARP in cancer cells that are trying to repair theDNA damage induced by temozolomide. The role ofPARP in DNA repair has not been fully elucidated andadditional roles for PARP in DNA damage signalling orrepair might explain the increased toxicity of combina-tion treatments57,58.

    Platinum chemotherapies.Cisplatin, carboplatinandoxaliplatinhave become three of the most commonly

    prescribed chemotherapeutic drugs used to treat solidcancers in patients59. Platinum resistance, either intrin-sic or acquired during cyclical treatment, is a majorclinical problem as additional agents that can be addedto therapy in order to circumvent tumour resistance donot currently exist.

    Platinum chemotherapy is now being tested with PARPinhibition in two clinical trials (TABLE 1). The rationale forcombining PARP inhibition with platinum chemotherapyis based on preclinical observations that PARP inhibitorspreferentially kill neoplastic cells andinduce completeor partial regression of a wide variety of human tumourxenografts in nude mice treated with platinum chemo-

    therapy6062. For example, ABT-888 (Abbott Laboratories,Chicago, Illinois, USA), a potent inhibitor of PARP1 andPARP2,has been shown to potentiate the regressionofestablished tumours induced by temozolomide, cisplatin,carboplatin or cyclophosphamide therapy in rodent ortho-topic and xenograft models63. However, as stated above,the full function of PARP in DNA repair is not clear57,58,and as a result the biological mechanisms of chemosensiti-zation of cancer cells to platinum chemotherapy by PARPinhibition remain to be resolved. Interestingly, as a mono-therapy in these preclinical models,ABT-888 exhibits nosignificant anticancer activity.

    DNA demethylating agents such as 2 -deoxy-5-azacytidine (decitabine; MGI Pharma, Bloomington,Minnesota, USA) have been combined with cisplatinor carboplatin to reverse drug resistance caused by the

    silencing of mismatch repair genes by hypermethylation.The toxicity of agents such as cisplatin depends at leastpartly on functional mismatch repair (FIG. 1). Preclinicaldata from xenograft models and translational studiesfrom drug-resistant cells and tissues that are mismatchrepair-deficient owing toMLH1hypermethylation havedemonstrated increased chemotherapeutic efficacywhen a demethylating agent is combined with platinumchemotherapy64,65. Decitabine is currently being testedin combination with carboplatin in a phase II clinicaltrial in patients with ovariancancer(see Decitabineand Carboplatin in Relapsed Ovarian Cancer in Furtherinformation).

    Table 1 | Ongoing clinical trials of small-molecule inhibitors of the DNA damage response and related signalling pathways

    Agent(company)

    Target moleculeor pathway

    Monotherapyor combinationtherapy agent(s)

    Phase of clinical trialplanned, ongoing orrecently completed

    Reference

    AZD-2281(Astra Zeneca)

    PARP GemcitabineCarboplatinTopotecan

    Monotherapy

    Phase IPhase IPhase I

    Phase II

    http://www.astrazenecaclinicaltrials.com/article/525925.aspx

    AG014699(Pfizer)

    PARP TemozolomideantibodyTemozolomide

    Phase I

    Phase II

    http://www.eddn.org/clinicalTr_caResUK.html

    INO-1001 (Inotek) PARP Temozolomide Phase I http://www.inotekcorp.com/content/ino-1001.asp

    BSI-201(Bipar Sciences)

    PARP MonotherapyGemcitabinecarboplatin

    Phase IPhase II

    http://www.biparsciences.com/BSI201.html

    ABT-888 (AbbottLaboratories)

    PARP Temozolomide Phase I NCT00526617

    TRC-102(Tracon Pharma)

    BER TemozolomidePemetrexed

    Phase IPhase I planned

    http://www.traconpharma.com/content/pipeline_overview.html

    Lomeguatrib(Astra Zeneca)

    MGMT IrinotecanTemozolomide

    Phase IPhase II

    http://www.astrazenecaclinicaltrials.com/article/525925.aspx

    O6-Benzylguanine MGMT Temozolomide Phase II http://clinicalstudies.info.nih.gov/cgi/detail.cgi?A_2006-C-0089.html

    Decitabine(MGI Pharma121)

    Hypermethylationof mismatchrepair genes

    Epirubicin,cisplatin,5-fluorouracilCarboplatin

    Phase I

    Phase II

    http://pfsearch.ukcrn.org.uk/StudyDetail.aspx?TopicID=&StudyID=2192

    XL844 (Exilixis122) CHK1, CHK2 Gemcitabine Phase I planned http://www.exelixis.com/pipeline_xl844.shtml

    The recent or current stage of development of clinical trials is indicated for individual compounds, which are grouped by molecular target. BER, base excisionrepair; CHK, checkpoint kinase; MGMT, O-6-methylguanine methyltransferase; PARP, poly(ADP-ribose) polymerase.

    R E V I E W S

    NATURE REVIEWS |CANCER VOLUME 8 |MARCH 2008 |197

    http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39176http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42374http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=10038http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39299http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=4292http://www.cancer.gov/cancertopics/types/ovarianhttp://www.astrazenecaclinicaltrials.com/article/525925.aspxhttp://www.astrazenecaclinicaltrials.com/article/525925.aspxhttp://www.eddn.org/clinicalTr_caResUK.htmlhttp://www.inotekcorp.com/content/ino-1001.asphttp://www.biparsciences.com/BSI201.htmlhttp://clinicaltrials.gov/ct/show/NCT00526617http://www.traconpharma.com/content/pipeline_overview.htmlhttp://www.traconpharma.com/content/pipeline_overview.htmlhttp://www.astrazenecaclinicaltrials.com/article/525925.aspxhttp://www.astrazenecaclinicaltrials.com/article/525925.aspxhttp://clinicalstudies.info.nih.gov/cgi/detail.cgi?A_2006-C-0089.htmlhttp://clinicalstudies.info.nih.gov/cgi/detail.cgi?A_2006-C-0089.htmlhttp://pfsearch.ukcrn.org.uk/StudyDetail.aspx?TopicID=&StudyID=2192http://pfsearch.ukcrn.org.uk/StudyDetail.aspx?TopicID=&StudyID=2192http://www.exelixis.com/pipeline_xl844.shtmlhttp://www.exelixis.com/pipeline_xl844.shtmlhttp://pfsearch.ukcrn.org.uk/StudyDetail.aspx?TopicID=&StudyID=2192http://pfsearch.ukcrn.org.uk/StudyDetail.aspx?TopicID=&StudyID=2192http://clinicalstudies.info.nih.gov/cgi/detail.cgi?A_2006-C-0089.htmlhttp://clinicalstudies.info.nih.gov/cgi/detail.cgi?A_2006-C-0089.htmlhttp://www.astrazenecaclinicaltrials.com/article/525925.aspxhttp://www.astrazenecaclinicaltrials.com/article/525925.aspxhttp://www.traconpharma.com/content/pipeline_overview.htmlhttp://www.traconpharma.com/content/pipeline_overview.htmlhttp://clinicaltrials.gov/ct/show/NCT00526617http://www.biparsciences.com/BSI201.htmlhttp://www.inotekcorp.com/content/ino-1001.asphttp://www.eddn.org/clinicalTr_caResUK.htmlhttp://www.astrazenecaclinicaltrials.com/article/525925.aspxhttp://www.astrazenecaclinicaltrials.com/article/525925.aspxhttp://www.cancer.gov/cancertopics/types/ovarianhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=4292http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39299http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=10038http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42374http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=39176
  • 8/11/2019 Dna Damage Helle

    6/12

    Synthetic lethality

    A genetic phenomenon in

    which the combination of two

    otherwise non-lethal

    mutations results in an

    inviable cell. Synthetic lethal

    phenotypes are indicative of

    an interaction between the

    products of the two mutant

    genes within the cell.

    It has been shown in preclinical models that the majorcisplatin intra-strand crosslinks formed in DNA are rec-ognized and repaired by the mammalian NER pathway66.One biological predictor of clinical outcome for patientswith completely resected non-small-cell lung cancerreceiving adjuvant cisplatin-based chemotherapy isexpression of the excision repair cross-complementationgroup 1 (ERCC1) protein67. An immunohistochemicalstudy of 761 operative specimens of non-small-cell lungcancer tissue taken before a proportion of the patients inthe study received adjuvant chemotherapy showed that asignificant benefit from cisplatin-based adjuvant chemo-therapy was associated with the absence of ERCC1 in56% of the samples studied. The results suggested thatpatients with completely resected non-small-cell lungcancer and ERCC1-negative tumours appeared tobenefit from adjuvant cisplatin-based chemotherapy,whereas patients with ERCC1-positive tumours did not.However, it should be noted that, among the patientswho did not receive adjuvant chemotherapy, those withERCC1-positive tumours survived significantly longer

    than those with ERCC1-negative tumours. It is thereforeconceivable from the results of this study that ERCC1might not represent a biomarker specific to cisplatin-based chemotherapy, but it might represent a marker ofprognosis and response to combination chemotherapy(not necessarily specific to cisplatin) in a poor-prognosissubset of patients, as has recently been demonstrated forp53overexpression in a similar patient subgroup68.

    Although it has been suggested that ERCC1 is apotential anticancer drug target, the protein has noknown enzymatic activity, making the means of regu-lating its activity harder to decipher. Pharmacologicalmodulation of ERCC1 might therefore be less desirablethan a greater understanding of the clinical relevance ofproteinprotein interactions within the NER machin-ery or between ERCC1 and other repair pathways, aspotential targets for improving the efficacy of platinumchemotherapies. For example, UCN-01(7-hydroxy-staurosporine) is an anticancer agent that potentiatescisplatin and carboplatin toxicity (shown in preclinicalmodels and a phase I clinical trial, respectively), whichhas been shown to interfere with the interaction ofERCC1 and another component of the NER pathway,xeroderma pigmentosum A (XPA), in vitro69.

    Attenuators of checkpoint signall ing.An alternativeapproach to modulating DNA repair activity and poten-

    tially improving the therapeutic index is to interfere withcell cycle checkpoint signalling. XL844(EXEL-9844) isa small-molecule inhibitor of CHK1 and CHK2. Thisdrug causes inhibition of cell-cycle arrest, progressiveDNA damage, inhibition of DNA repair and, ultimately,tumour cell apoptosis in cancer cells grown in vitro70,although the outcome of inhibiting CHK1 and CHK2in general can vary in a cell type-dependent manner.Although concerns have been raised about the potentialtoxicity to normal cells of an approach which inhibitsboth CHK1 and CHK2 kinases71, preclinical data havesuggested that intermittent dosing with XL844 incombination with the deoxycytidine analoguegemcitabine

    is well tolerated by female athymic nude mice used asa xenograft model70. XL844 is currently being testedin a clinical trial (NCT00475917) in combination withgemcitabine, which normally causes cell-cycle arrest andapoptosis by its incorporation into DNA.

    Two kinases from the phosphatidylinositol 3-kinase(PI3K)-related protein kinase (PIKKs) family, ATM andATR, are central to cellular responses to DSBs. Whenactivated, ATM and ATR phosphorylate a multitude ofproteins, which initiate a cascade that induces cell-cyclearrest and facilitatesDNA repair. An inhibitor of ATMkinase activity, KU55933 (AstraZeneca), is currentlyin preclinical development. The rationale behind theclinical use of ATM inhibitors depends on the premisethat ATM inhibition should improve the therapeuticindex by hypersensitizing tumour cells to agents thatcause DSBs. Here again, the feasibility of inhibitingATM kinase in patients will depend on the level oftoxicity that such agents cause in normal tissues whenthey are used in combination with ionizing radiation orcytotoxic chemotherapy.

    Radiosensitizers. DNA-dependent protein kinase(DNAPK, also known as PRKDC), a member of the PIKKfamily, is important for DSB repair by non-homologousend joining following ionizing radiation72. Cells defectivein DNAPK are highly sensitive to ionizing radiation73indicating that inhibition of DNAPK might sensitizetumours to radiation treatment. Wortmannin, a knowninhibitor of PIKKs at low nanomolar concentrations,has antiproliferative effects and is a radiosensitizer inpreclinical models74, but is unsuitable for clinical appli-cations owing to its inherent toxicity and instability incells75. Other small molecules that reversibly inhibitDNAPK kinase activity at low micromolar concentra-tions have been synthesized. They are currently in tran-sition from late preclinical development to early clinicaltrials. In particular, NU7441 (REF.76)has been shownto sensitize cells to topoisomerase II poisons and canalso function as a radiosensitizer in a manner consistentwith DNAPK inhibition77.

    DNA repair inhibitors as monotherapy

    As discussed above, most of the current small-moleculeinhibitors of DNA repair have so far been tested in earlyclinical trials as sensitizers of tumour cells to chemo-therapy. However, DNA damage also occurs spontane-ously in cells in the absence of treatments and DNA

    repair pathways are therefore essential for the survivalof untreated cells. As several cancers are defective inDNA damage response and repair pathways (TABLE 2),the concept of synthetic lethal interactions can be used toadvocate the use of DNA repair inhibitors as monothera-pies (FIG. 2). DNA repair is an ideal target for inhibition incancer cells as the inhibitors should be exclusively toxicto cancer cells and therefore be associated with minimalside effects for patients (BOX 2).

    Indeed, DNA repair inhibitors have been demon-strated to work as single agents in patients with DNArepair-defective tumours. The most notable example sofar is the use of PARP inhibitors to treat patients with

    R E V I E W S

    198 |MARCH 2008 |VOLUME 8 www.nature.com/reviews/cancer

    http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2067http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7157http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42305http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7507http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=469731http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=41213http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=41213http://clinicaltrials.gov/ct/show/NCT00475917jsessionid=94D09AADF5BBF70FC2857F92FBE6C03A?order=12http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5591http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5591http://clinicaltrials.gov/ct/show/NCT00475917jsessionid=94D09AADF5BBF70FC2857F92FBE6C03A?order=12http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=41213http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=469731http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7507http://www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42305http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7157http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2067
  • 8/11/2019 Dna Damage Helle

    7/12

    Table 2 | Synthetic lethal interactions of DNA repair and cell cycle checkpoint genes implicated in cancer by pathway

    Protein Syndrome Primary cancers Biomarker SLIs S. cerevisiaehomologue

    SLIs in S.cerevisiae (n)

    HR

    BRCA1, BRCA2 Breast, ovarian112, 135 PARP1 (REFS 78,79)

    RAD54B NHL, colon cancer113 PARP1 (REF. 87) rdh54

    RAD51B (RAD51L1) Lipoma, uterineleiomyoma114 PARP1 (REF. 87) rad51 31

    CtIP (RBBP8) Colorectal cancer115 sae2 5

    NHEJ

    MRE11 Ataxiatelangiectasia-like disorder

    Colorectal cancer108 IR sensitivity mre11 40

    LIG4 LIG4 Leukaemia116 IR sensitivity lig4

    Artemis (DCLRE1C) Omenn Lymphoma117 pso2

    MMR

    MSH2, MLH1, MSH6,PMS1, PMS2,MLH3

    Hereditary nonpolyposiscolorectal cancer118120

    Microsatelliteinstability

    msh2, mlh1,msh6, pms1, mlh3

    3

    RecQ

    BLM Bloom Various121

    Increased SCE sgs1 43WRN Werner Various122 Increased

    telomeric SCE sgs1 43

    RECQL4 RothmundThomson

    Skin basal and sqamouscell, osteosarcoma122

    sgs1 43

    Damage signalling

    ATM Ataxia-telangiectasia

    Leukaemia123 IR sensitivity PARP1(REFS 124,125),FANC89

    tel1 2

    NBS1 Nijmegenbreakage

    Various126 IR sensitivity xrs2 30

    p53 LiFraumeni Various127 -

    CHK2 LiFraumeni Various128 dun1/rad53 37

    NERXPA, XPC, DDB1,ERCC4,ERCC5, POLH

    XP Skin cancers129 UV sensitivity rad14, rad4,rad1, rad2, rad30

    11

    ERCC2,ERCC3 XP, Cockayne,trichothio-dystrophy

    Skin cancers129 rad25, rad3 8

    ERCC1 Cerebro-oculo-facio-skeletal

    Squamous cell carcinoma,head and neck130

    rad10 6

    FA

    FANCA,FANCC,FANCD2, FANCE,FANCG

    Fanconianaemia

    Various131 Impaired FANCD2ubiqutination131

    ATM89

    FANCB, FANCF,FANCL Fanconianaemia

    Various131 Impaired FANCD2ubiqutination131

    BRIP1 Fanconianaemia

    Various131

    FANCM Fanconianaemia

    Various131 Impaired FANCD2ubiqutination131

    mph1 1

    BER

    POLB Various132 pol4

    FEN1 Various133 rad27 104

    Synthetic lethalities observed in mammalian cells and the number of synthetic lethal interactions (SLIs) for their Saccharomyces cervisiaehomologues are shown.The genes showing SLIs with DNA repair genes can potentially be used as targets for novel drugs that then may selectively kill cancer cells in monotherapy.A complete list of the SLIs in S. cervisiaecan be found in SupplementaryinformationS1(table). ATM, ataxia telangiectasia mutated; BER, base-excision repair;CHK, checkpoint kinase; ERCC, excision repair cross-complementation; FA, Fanconi anaemia-mediated repair; FANC, Fanconi anemia, complementation group;FEN1, flap structure-specific endonuclease 1; HR, homologous recombination; IR, infrared; MLH, mutL homologue; MMR, mismatch repair; MSH, mutShomologue; NER, nucleotide-excision repair; NHEJ, non-homologous end joining; PARP, poly(ADP-ribose) polymerase; RecQ, RecQ-mediated repair; SCE, sisterchromatid e xchange; UV, ultraviolet; XP, xeroderma pigme ntosum.

    R E V I E W S

    NATURE REVIEWS |CANCER VOLUME 8 |MARCH 2008 |199

    http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=25788http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5890http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=3981http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=64421http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2956http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2956http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5378http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5395http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5395http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=27030http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=9401http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7508http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=1642http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2072http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2073http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2073http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5429http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2068http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2071http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2071http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2175http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2176http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2176http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2177http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2178http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2189http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2187http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2188http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=55120http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=55120http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=83990http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=57697http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5423http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2237http://www.nature.com/nrc/journal/v8/n3/suppinfo/nrc2342.htmlhttp://www.nature.com/nrc/journal/v8/n3/suppinfo/nrc2342.htmlhttp://www.nature.com/nrc/journal/v8/n3/suppinfo/nrc2342.htmlhttp://www.nature.com/nrc/journal/v8/n3/suppinfo/nrc2342.htmlhttp://www.nature.com/nrc/journal/v8/n3/suppinfo/nrc2342.htmlhttp://www.nature.com/nrc/journal/v8/n3/suppinfo/nrc2342.htmlhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2237http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5423http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=57697http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=83990http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=55120http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2188http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2187http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2189http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2178http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2177http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2176http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2175http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2071http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2068http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5429http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2073http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2072http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=1642http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7508http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=9401http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=27030http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5395http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5378http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2956http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=64421http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=3981http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5890http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=25788
  • 8/11/2019 Dna Damage Helle

    8/12

    |

    Targeted monotherapyCancer mutation

    Replication lesion

    Survival

    a Initial cancer mutation

    Lethal

    b Inhibitor of Top3 in cancer cells

    Survival

    c Drug-resistant mutation d Second therapy in resistant cells

    Lethal

    SAE2mre11

    SGS1

    TOP3 mre11

    SGS1

    TOP3

    sgs1

    Gene function Drug-resistant mutation

    Monotherapy Mutation Monotherapymre11 mre11SAE2

    sgs1

    inherited breastand ovarian cancers that lack wild-typecopies of the BRCA1or BRCA2genes78,79. BRCA1- andBRCA2-mutated cells are defective in HR repair80,81and show extensive replication-associated lesions82,83.These recombination-defective cells are 1001,000-fold more sensitive to PARP inhibitors than are theheterozygote or the wild-type cell lines, indicatingtheir potential to be exploited as specific treatments ofBRCA1-or BRCA2-defective tumours78,79. One explana-tion for this sensitivity is that PARP inhibitors inducesingle-strand breaks that can result in DSBs as a resultof stalled replication forks. Such lesions would normallybe repaired by HR, but this is prohibited in BRCA1- orBRCA2-deficient cancer cells79,8486. PARP activity is alsorequired for the actions of CHFR(checkpoint proteinwith forkhead-associated and ring finger domains)58andthe reactivation of stalled replication forks57. These func-tions might also explain the hypersensitivity to PARPinhibitors of recombination-defective cells. Translationof these observations has led to phase II clinical trials of

    monotherapy using the PARP inhibitor AZD2281(AstraZeneca) currently recruiting patients with breastand ovarian cancer who harbour mutations in BRCA1orBRCA2genes. A separate phase II trial with the PARP1inhibitor AG014699 (Pfizer GRD) is due to open torecruitment of known carriers of BRCA1or BRCA2mutations with locally advanced or metastatic cancers ofthe breast or ovaries. It should be noted however that notall patients with mutations in BRCA1or BRCA2respondto PARP inhibitors as a monotherapy. The reasons forthis are currently under investigation. Cells that aredefective in recombination-related proteins other thanBRCA1 or BRCA2, such as RAD51, RAD54, XRCC2,

    XRCC3, DSS1 (also known as SHFM1), replicationprotein A1 (RPA1), ATM, ATR, CHK1, CHK2, NBS1(also known as NBN) and components of the Fanconianaemia repair pathway, also show increased sensitivityto PARP inhibition79,87,88. This suggests that PARP inhibi-tors might also be suitable in treating several types oftumours with defects in HR.

    Another synthetic lethal interaction has recentlybeen discovered between the Fanconi anaemia repairpathway and ATM. Two pancreatic tumour lines defec-tive in the Fanconi anaemia pathway were more sensitiveto the ATM inhibitor KU-55933 than isogenic controlcells89. This finding provides a rationale for studyingATM inhibitors in the treatment of Fanconi anaemiarepair-defective pancreatic cancer.

    Finding new synthetic lethal DNA repair-based partner-

    ships.As mutations in checkpoint and DNA repairpathways are associated with cancer (TABLE 2), it shouldbe straightforward to exploit DNA repair inhibitors

    for the treatment of tumours carrying specific defectsin DNA repair or damage signalling (FIG. 2). Genome-wide screens in model organisms, such as the bud-ding yeast Saccharomyces cerevisiae, have helped toidentify protein interaction networks, which serve toelucidate protein functions within highly complex cel-lular processes. Such knowledge can prove extremelyuseful in identifying suitable targets for monotherapy,but perhaps more significantly those that could be usedin combination therapies. We have compiled a list ofreported mutations in DNA damage response genesfound in human tumours and homologous syntheticlethal interactions that have been demonstrated in

    Figure 2 |Synthetic lethal interactions to identify molecular targets for inhibitors of DNA repair. A mutation in a

    single tumour suppressor gene can result in genetic instability that can accelerate tumour progression90. Here, we use

    synthetic lethal interactions in Saccharomyces cerevisiaeto illustrate a hypothetical scenario. a| We start with an

    Mre11mutation that is found in mismatch repair-defective tumours108. b| The yeast mre11is synthetic lethal with

    topoisomerase III (Top33)109. If the synthetic lethal interaction is conserved from yeast to man, inhibitors of Top3 would,

    in theory, kill Mre11-mutated tumour cells. Top3-mutated yeast cells are viable, albeit with genetic instability110,

    and thus Top3 inhibitors may specifically kill Mre11-mutated cancer cells while being tolerated in normal cells.

    c| Replication repair networks are complex and an additional mutation might override synthetic lethal interactions. In

    yeast, an additional mutation in sgs1, upstream of the hypothetically targetedTOP3pathway, reverses the synthetic

    lethal phenotype

    109

    . Thus, in this scenario, an additional tumour mutation in the human homologue of SGS1, BLM, mightresult in resistance to Top3 inhibitors. d| sae2is in turn synthetic lethal with sgs1in yeast111. If this synthetic lethal

    interaction is conserved, the human SAE2homologue CtIP (also known as retinoblastoma binding protein 8 (RBBP8))

    might, in theory, then be targeted as a second-line therapy in Top3 inhibitor-resistant tumours that gained resistance

    owing to a BLM mutation. In summary, understanding of DNA repair networks is important to identify pathways to

    target and to circumvent resistance mechanisms.

    R E V I E W S

    200 |MARCH 2008 |VOLUME 8 www.nature.com/reviews/cancer

    http://www.cancer.gov/cancertopics/types/breasthttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=672http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=675http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=55743http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5888http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7516http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7517http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7979http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=6117http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=4683http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=855264http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=855264http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=850935http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=855228http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=852700http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5932http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5932http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=852700http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=855228http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=850935http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=855264http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=4683http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=6117http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7979http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7517http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=7516http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=5888http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=55743http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=675http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=672http://www.cancer.gov/cancertopics/types/breast
  • 8/11/2019 Dna Damage Helle

    9/12

    Biomarkers

    A molecule or substance

    whose detection indicates a

    particular disease state or

    treatment response.

    Hypoxia

    A subnormal concentration of

    oxygen. In cancer tissue,

    hypoxia is often the result of

    abnormal vasculature.

    S. cerevisiae(SupplementaryinformationS1(table)).

    This information could be used to identify proteinsencoded by the human homologues of genes showingsynthetic lethal interactions that may represent goodtargets for specific treatment of cancers carrying muta-tions in DNA repair genes. Unfortunately, the poorlydefined biochemical properties of these proteins arestill a significant barrier to the exploitation of theseproteins as potential targets for drug intervention inthe immediate future.

    A further limitation to exploiting these pathwaysis the lack of reliable biomarkersto aid the selection ofpatients that might respond to such treatments. This isparticularly important as cancers in patients that have no

    DNA repair defect will not respond. The most reliable

    biomarkers will probably be those that identify loss ofspecific post-translational modifications present in theDNA damage response and repair pathways, or thosethat indicate increased activity of the targeted pathway.

    Future directions

    Current chemotherapy regimens demonstrate that pro-duction of excessive replication lesions is a successfulmeans of killing cancer cells. It has been observed thattumour cells themselves exhibit a high level of endogenousreplication lesions that result in genetic instability32,33,90.In theory, DNA repair inhibitors could be used to impairthe repair of replication lesions that are present in tumourcells and convert them into fatal replication lesions thatspecifically kill cancer cells. For example, there is evidencethat the increased expression or activity of oncogenes caninduce replication stress32,33,91,92. In such tumours it mightbe possible to use DNA repair inhibitors to make existingcancer-specific replication lesions more toxic, resultingin fatal replication lesions selectively killing oncogene-expressing cancer cells32,33,91972(BOX 3).

    More advanced cancers are exposed to another sourceof replication stress owing to the tumour microenviron-ment. Tumours are often hypoxic, which has been shownto disrupt DNA synthesis98. These conditions cause repli-cation lesions that activate the ATM- and ATR-mediatedcheckpoint response99101. Furthermore, DNA repair is

    downregulated in hypoxic cells102, which cumulativelycontributes to the genetic instability observed in thesecells103,104. In hypoxic cancer cells, therefore, inhibitors ofthe checkpoint response could prove to be more efficientthan inhibitors of DNA repair105.

    Conclusions

    The potential of inhibitors of DNA repair in the futureof cancer therapy is starting to become apparent.Although selective inhibition of DNA repair pathwayscan be used to enhance current chemotherapy, themost attractive use of DNA repair inhibitors might bein using cancer defects for selective cell killing. DNA

    Box 2 | Advantages and limitations using DNA repair inhibitors as single agents in treatment of cancers

    DNA repair inhibitors can exploit tumour-specific defects in checkpoint signalling and DNA repair to convert

    endogenous DNA lesions into fatal replication lesions that selectively kill tumour cells.

    A general problem for novel cancer therapies is that they are not sufficiently efficient at killing cancer cells to

    replace current cytotoxic therapies. As a result, some enzyme inhibitors (that do not target DNA repair) have failed

    in late clinical trial development owing to a general lack of anti-tumour efficacy. Inhibition of DNA repair amplifies

    toxic replication-associated DNA lesions that directly result in cell death. DNA repair inhibitors might therefore be

    highly efficient at killing tumours.

    Cross-talk between DNA repair pathways in normal cells minimizes side effects from the inhibition of a single DNA

    repair pathway.

    Tumour inactivation of DNA damage signalling and DNA repair are often relatively early events during carcinogenesis,

    suggesting that non-toxic DNA repair inhibitors may be considered in the treatment of patients with pre-malignant or

    early neoplastic lesions, such as those arising in patients with inherited mutations inBRCA1orBRCA2genes, or

    intestinal lesions in patients with defects inMLH1andMSH2genes or their methylation status.

    Cross-talk between DNA repair pathways, such as the cross-talk between homologous recombination and non-

    homologous end joining in the repair of DNA double-strand breaks or the cross-talk between base-excision repair,

    alkyltransferases and DNA dioxygenases in the repair of alkylation damage, is likely to result in acquisition of

    resistance mechanisms in tumours, which is a limitation for killing more advanced tumours.

    Box 3 | Targetting oncogene-induced replication stress

    The transformation of normal cells to a cancerous state is often initiated by the

    activation of oncogenes, which provide excessive growth signals106. It was recently

    shown that such oncogene activation can induce replication-associated DNA

    lesions32,33,91,92, including increased replication origin firing, re-replication and

    impaired fork progression32,107. These replication-associated lesions trigger a cell-cycle

    checkpoint response that stops the proliferation of pre-cancerous cells early during

    neoplastic transformation32,33,9193. This involves checkpoint proteins, such as p53 and

    CHK2, that can induce apoptosis or senescence94,95,

    Genes encoding proteins in the checkpoint pathways are often mutated during

    cancer development96, allowing cells to evade checkpoints and continue to

    proliferate. Taken together, these observations suggest that a key feature of cancer

    cells that overexpress oncogenes might be higher levels of endogenous replication-associated lesions than those present in normal cells. This in turn would contribute to

    genetic instability97that would assist the tumour to induce the genetic changes

    required for continuing transformation to malignant phenotypes90. More importantly,

    the replication lesions caused by oncogene activation have been found to resemble

    those produced by anticancer treatments32, and, like the latter, these lesions would

    require repair for the cancer cells to survive. We therefore propose that future DNA

    repair inhibitors should be used to make existing cancer-specific replication lesions

    more toxic, resulting in fatal replication lesions selectively killing oncogene-

    expressing cancer cells. The nature of the replication lesions that are produced

    following chemotherapy or oncogene-induced stress are poorly understood.

    Although several replication repair pathways have been identified, we currently have

    little information regarding their complex interplay. Indeed, more intense basic

    research is required in this area to identify novel anticancer targets.

    R E V I E W S

    NATURE REVIEWS |CANCER VOLUME 8 |MARCH 2008 |201

    http://www.nature.com/nrc/journal/vaop/ncurrent/suppinfo/nrc2342.htmlhttp://www.nature.com/nrc/journal/vaop/ncurrent/suppinfo/nrc2342.htmlhttp://www.nature.com/nrc/journal/vaop/ncurrent/suppinfo/nrc2342.htmlhttp://www.nature.com/nrc/journal/vaop/ncurrent/suppinfo/nrc2342.htmlhttp://www.nature.com/nrc/journal/vaop/ncurrent/suppinfo/nrc2342.htmlhttp://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=4436http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=4436http://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=4436http://www.nature.com/nrc/journal/vaop/ncurrent/suppinfo/nrc2342.html
  • 8/11/2019 Dna Damage Helle

    10/12

    1. Hsiang, Y. H., Lihou, M. G. & Liu, L. F. Arrest of

    replication forks by drug-stabilized topoisomerase I

    DNA cleavable complexes as a mechanism of cell

    killing by camptothecin. Cancer Res.49, 50775082

    (1989).

    2. Markovits, J. et al.Topoisomerase II-mediated DNA

    breaks and cytotoxicity in relation to cell

    proliferation and the cell cycle in NIH 3T3 fibroblasts

    and L1210 leukemia cells. Cancer Res.47,

    20502055 (1987).

    3. Ikegami, S. et al.Aphidicolin prevents mitotic cell

    division by interfering with the activity of DNA

    polymerase-. Nature275, 458460 (1978).4. Bianchi, V., Pontis, E. & Reichard, P. Changes of

    deoxyribonucleoside triphosphate pools induced by

    hydroxyurea and their relation to DNA synthesis.

    J. Biol. Chem.261, 1603716042 (1986).5. Lundin, C. et al.Different roles for nonhomologous

    end joining and homologous recombination following

    replication arrest in mammalian cells. Mol. Cell Biol.

    22, 58695878 (2002).

    6. Saintigny, Y. et al.Characterization of homologous

    recombination induced by replication inhibition inmammalian cells. EMBO J.20, 38613870 (2001).

    7. Swann, P. F. et al.Role of postreplicative DNA

    mismatch repair in the cytotoxic action of

    thioguanine. Science273, 11091111 (1996).

    8. Wang, J. C. Cellular roles of DNA topoisomerases: a

    molecular perspective. Nature Rev. Mol. Cell Biol.3,

    430440 (2002).

    9. Painter, R. B. & Cleaver, J. E. Repair replication in

    HeLa cells after large doses of x-irradiation. Nature

    216, 369370 (1967).

    10. Canman, C. E. et al.Activation of the ATM kinase by

    ionizing radiation and phosphorylation of p53.

    Science281, 16771679 (1998).

    11. Falck, J., Mailand, N., Syljuasen, R. G., Bartek, J. &

    Lukas, J. The ATMChk2Cdc25A checkpoint

    pathway guards against radioresistant DNA synthesis.

    Nature410, 842847 (2001).

    This paper describes the molecular mechanism by

    which ATM rapidly inactivates DNA synthesis

    following ionizing radiation.12. Cliby, W. A. et al.Overexpression of a kinase-inactive

    ATR protein causes sensitivity to DNA-damaging

    agents and defects in cell cycle checkpoints. EMBO J.

    17, 159169 (1998).

    13. Taylor, A. M. et al.Ataxia telangiectasia: a human

    mutation with abnormal radiation sensitivity. Nature

    258, 427429 (1975).

    14. Sargent, R. G., Brenneman, M. A. & Wilson, J. H.

    Repair of site-specific double-strand breaks in a

    mammalian chromosome by homologous and

    illegitimate recombination. Mol. Cell Biol.17,

    267277 (1997).15. Arnaudeau, C., Lundin, C. & Helleday, T. DNA double-

    strand breaks associated with replication forks are

    predominantly repaired by homologous

    recombination involving an exchange mechanism in

    mammalian cells.J. Mol. Biol.307, 12351245

    (2001).

    16. Sharma, R. A. & Dianov, G. L. Targeting base excision

    repair to improve cancer therapies. Mol. Aspects

    Med.28, 345374 (2007).

    17. Huang, J. C., Svoboda, D. L., Reardon, J. T. & Sancar, A.

    Human nucleotide excision nuclease removes thymine

    dimers from DNA by incising the 22nd

    phosphodiester bond 5and the 6th phosphodiesterbond 3to the photodimer. Proc. Natl Acad. Sci. USA89, 36643668 (1992).

    18. Sugasawa, K. et al.A multistep damage recognition

    mechanism for global genomic nucleotide excision

    repair. Genes Dev.15, 507521 (2001).

    19. Sedgwick, B. Repairing DNA-methylation damage.

    Nature Rev. Mol. Cell Biol.5, 148157 (2004).

    20. Lindahl, T., Demple, B. & Robins, P. Suicide

    inactivation of the E. coliO6-methylguanine-DNA

    methyltransferase. EMBO J.1, 13591363 (1982).21. Duncan, T. et al.Reversal of DNA alkylation damage

    by two human dioxygenases. Proc. Natl Acad. Sci.

    USA 99, 1666016665 (2002).

    22. Karran, P. & Marinus, M. G. Mismatch correction at

    O6-methylguanine residues in E. coliDNA. Nature

    296, 868869 (1982).23. Yoshioka, K., Yoshioka, Y. & Hsieh, P. ATR kinase

    activation mediated by MutSand MutLin responseto cytotoxic O6-methylguanine adducts. Mol. Cell22,

    501510 (2006).

    24. Fram, R. J., Cusick, P. S., Wilson, J. M. & Marinus,

    M. G. Mismatch repair of cis-diamminedichloroplatin

    um(II)-induced DNA damage. Mol. Pharmacol.28,

    5155 (1985).25. Masutani, C., Kusumoto, R., Iwai, S. & Hanaoka, F.

    Mechanisms of accurate translesion synthesis by

    human DNA polymerase . EMBO J.19, 31003109(2000).

    26. Vaisman, A., Masutani, C., Hanaoka, F. & Chaney,

    S. G. Efficient translesion replication past oxaliplatin

    and cisplatin GpG adducts by human DNA

    polymerase . Biochemistry39, 45754580 (2000).27. Fukui, T. et al.Distinct roles of DNA polymerases

    and at the replication fork inXenopusegg extracts.Genes Cells9, 179191 (2004).

    28. Pursell, Z. F., Isoz, I., Lundstrom, E. B., Johansson, E.& Kunkel, T. A. Yeast DNA polymerase participatesin leading-strand DNA replication. Science317,

    127130 (2007).

    29. Lehmann, A. R. Translesion synthesis in mammalian

    cells. Exp. Cell Res.312, 26732676 (2006).30. Sorensen, C. S. et al.The cell-cycle checkpoint kinase

    Chk1 is required for mammalian homologous

    recombination repair. Nature Cell Biol.7, 195201

    (2005).

    31. Kastan, M. B. & Bartek, J. Cell-cycle checkpoints and

    cancer. Nature432, 316323 (2004).

    32. Bartkova, J. et al.Oncogene-induced senescence is

    part of the tumorigenesis barrier imposed by DNA

    damage checkpoints. Nature444, 633637

    (2006).

    33. Di Micco, R. et al.Oncogene-induced senescence is a

    DNA damage response triggered by DNA hyper-

    replication.Nature444, 638642 (2006).

    References 32 and 33 show that oncogenes induce

    replication lesions in premalignant cancer cells

    that in turn activate senescence as a tumour

    barrier.34. Arnaudeau, C., Tenorio Miranda, E., Jenssen, D. &

    Helleday, T. Inhibition of DNA synthesis is a potent

    mechanism by which cytostatic drugs induce

    homologous recombination in mammalian cells.

    Mutat. Res. DNA Repair461, 221228 (2000).

    35. Helleday, T., Lo, J., van Gent, D. C. & Engelward, B. P.

    DNA double-strand break repair: From mechanistic

    understanding to cancer treatment. DNA Repair

    (Amst.)6, 923935 (2007).

    36. Patel, K. J. & Joenje, H. Fanconi anemia and DNA

    replication repair. DNA Repair (Amst.)6, 885890

    (2007).

    37. Hanada, K. et al.The structure-specific endonuclease

    Mus81 contributes to replication restart by

    generating double-strand DNA breaks. Nature Struct.

    Mol. Biol.14, 10961104(2007).

    38. Karow, J. K., Constantinou, A., Li, J. L., West, S. C.

    & Hickson, I. D. The Blooms syndrome gene

    product promotes branch migration of hollidayjunctions. Proc. Natl Acad. Sci. USA 97,

    65046508 (2000).

    39. Lebel, M., Spillare, E. A., Harris, C. C. & Leder, P.

    The Werner syndrome gene product co-purifies with the

    DNA replication complex and interacts with PCNA and

    topoisomerase I.J. Biol. Chem.274, 3779537799

    (1999).

    40. Constantinou, A. et al.Werners syndrome protein

    (WRN) migrates Holliday junctions and co-localizes with

    RPA upon replication arrest. EMBO Rep.1, 8084

    (2000).

    41. Wu, L. & Hickson, I. D. DNA helicases required for

    homologous recombination and repair of damaged

    replication forks.Annu. Rev. Genet.40, 279306

    (2006).42. Niedzwiedz, W. et al.The Fanconi anaemia gene FANCC

    promotes homologous recombination and error-prone

    DNA repair. Mol. Cell15, 607620 (2004).

    43. Wu, L. & Hickson, I. D. The Blooms syndrome

    helicase suppresses crossing over during homologousrecombination. Nature426, 870874 (2003).

    44. Chen, X. B. et al.Human Mus81-associated

    endonuclease cleaves Holliday junctions in vitro. Mol.

    Cell8, 11171127 (2001).

    45. Hinz, J. M., Nham, P. B., Urbin, S. S., Jones, I. M. &

    Thompson, L. H. Disparate contributions of the

    Fanconi anemia pathway and homologous

    recombination in preventing spontaneous

    mutagenesis. Nucleic Acids Res.35, 37333740

    (2007).

    46. Thompson, L. H. Strategies for cloning mammalian

    DNA repair genes. Methods Mol. Biol.113, 5785

    (1999).

    47. Chabner, B. A. & Roberts, T. G. Jr. Chemotherapy and

    the war on cancer. Nature Rev. Cancer5, 6572

    (2005).

    48. Stevens, M. F. et al.Antitumor activity and

    pharmacokinetics in mice of 8-carbamoyl-3-methyl-

    repair inhibitors that exploit tumour mutations inDNA repair pathways to convert spontaneous DNAlesions into fatal replication lesions might represent themost direct means of finding selective treatments forcertain cancers. This type of therapy is highly advan-tageous when compared with current chemotherapyas it is likely to produce minimal side effects whileresulting in highly toxic lesions that should activelytrigger cell death in cancer cells. A potential limita-tion of this approach is that it is likely to be confinedto DNA repair-defective tumours and that resistancemechanisms might develop.

    A more challenging treatment strategy is the inhibi-tion of the repair of tumour-specific replication lesionsand conversion of these into fatal lesions. Replicationstress appears to be present in a majority of tumoursduring at least one stage of carcinogenesis. Thus, theconversion of replication stress into fatal replication

    lesions could potentially be used to target a widerange of tumours. As we are still unaware of the exactnature of the replication lesions that are induced bymany traditional chemotherapies, there is still con-siderable work to be done in characterizing tumour-specific lesions to target cancers. Basic research intounderstanding the nature of toxic replication lesions,as well as obtaining a more complete picture of allDNA repair pathways and their interplay, is crucialfor the future of DNA repair inhibitors as single agentsin cancer therapy.

    In summary, cancer cells are potentially exposedto unusually high levels of replication stress and endo-genous DNA damage during cancer development. Afuture challenge will be to identify and characterizeforms of replication lesions occurring during carcino-genesis and neoplastic progression that may be exploitedfor selective therapy.

    R E V I E W S

    202 |MARCH 2008 |VOLUME 8 www.nature.com/reviews/cancer

  • 8/11/2019 Dna Damage Helle

    11/12

    imidazo[5,1-d]-1,2,3,5-tetrazin-4(3H)-one (CCRG

    81045; M & B 39831), a novel drug with potential

    as an alternative to dacarbazine. Cancer Res.47,

    58465852 (1987).

    49. Stupp, R. et al.Radiotherapy plus concomitant and

    adjuvant temozolomide for glioblastoma. N. Engl. J.

    Med.352, 987996 (2005).

    References 48 and 49 demonstrate the timescale

    required from drug discovery in the laboratory to

    the demonstration of improved survival in patients

    in a randomized phase III clinical trial, in this case

    for temozolomide as a radiosensitizer.50. Dolan, M. E. & Pegg, A. E.O6-Benzylguanine and its

    role in chemotherapy. Clin. Cancer Res.3, 837847

    (1997).

    51. Gerson, S. L., Berger, N. A., Arce, C., Petzold, S. J. &

    Willson, J. K. Modulation of nitrosourea resistance in

    human colon cancer by O6-methylguanine. Biochem.

    Pharmacol.43, 11011107 (1992).

    52. Middleton, M. R. & Margison, G. P. Improvement of

    chemotherapy efficacy by inactivation of a DNA-repair

    pathway. Lancet Oncol.4, 3744 (2003).

    53. Ranson, M. et al.Lomeguatrib, a potent inhibitor of

    O6-alkylguanine-DNA-alkyltransferase: phase I safety,

    pharmacodynamic, and pharmacokinetic trial and

    evaluation in combination with temozolomide in

    patients with advanced solid tumors. Clin. Cancer

    Res.12, 15771584 (2006).

    54. Quinn, J. A. et al.Phase I trial of temozolomide plus

    O6-benzylguanine for patients with recurrent or

    progressive malignant glioma.J. Clin. Oncol.23,

    71787187 (2005).

    55. Khan, O. & Middleton, M. R. The therapeutic potential

    of O6-alkylguanine DNA alkyltransferase inhibitors.

    Expert Opin. Investig. Drugs16, 15731584 (2007).

    56. Durkacz, B. W., Omidiji, O., Gray, D. A. & Shall, S.

    (ADP-ribose)nparticipates in DNA excision repair.

    Nature283, 593596 (1980).

    This study was first to demonstrate that PARP

    inhibitors can be used to increase the toxicity of

    DNA-damaging agents.57. Yang, Y. G., Cortes, U., Patnaik, S., Jasin, M. & Wang,

    Z. Q. Ablation of PARP-1 does not interfere with the

    repair of DNA double-strand breaks, but

    compromises the reactivation of stalled replication

    forks. Oncogene23, 38723882 (2004).

    58. Ahel, I.et al.Poly(ADP-ribose)-binding zinc finger

    motifs in DNA repair/checkpoint proteins. Nature

    451, 8185 (2008).

    59. Rosenberg, B., VanCamp, L., Trosko, J. E. & Mansour,

    V. H. Platinum compounds: a new class of potent

    antitumour agents. Nature222, 385386 (1969).

    60. Kubo, S. et al.Participation of poly(ADP-ribose)polymerase in the drug sensitivity in human lung

    cancer cell lines.J. Cancer Res. Clin. Oncol.118,

    244248 (1992).

    61. Miknyoczki, S. J. et al.Chemopotentiation of

    temozolomide, irinotecan, and cisplatin activity by

    CEP-6800, a poly(ADP-ribose) polymerase inhibitor.

    Mol. Cancer Ther.2, 371382 (2003).

    62. Robins, H. I. et al.Phase I trial of intravenous

    thymidine and carboplatin in patients with advanced

    cancer.J. Clin. Oncol.17, 29222931 (1999).

    63. Donawho, C. K. et al.ABT-888, an oral ly active

    poly(ADP-ribose) polymerase inhibitor that

    potentiates DNA-damaging agents in preclinical

    tumor models. Clin. Cancer Res.13, 27282737

    (2007).

    64. Gifford, G., Paul, J., Vasey, P. A., Kaye, S. B. &

    Brown, R. The acquisition of hMLH1 methylation in

    plasma DNA after chemotherapy predicts poor

    survival for ovarian cancer patients. Clin. Cancer Res.

    10, 44204426 (2004).65. Plumb, J. A., Strathdee, G., Sludden, J., Kaye, S. B. &

    Brown, R. Reversal of drug resistance in human tumor

    xenografts by 2-deoxy-5-azacytidine-induceddemethylation of the hMLH1 gene promoter. Cancer

    Res.60, 60396044 (2000).

    66. Rabik, C. A. & Dolan, M. E. Molecular mechanisms of

    resistance and toxicity associated with platinating

    agents. Cancer Treat. Rev.33, 923 (2007).

    67. Olaussen, K. A. et al.DNA repair by ERCC1 in non-

    small-cell lung cancer and cisplatin-based adjuvant

    chemotherapy. N. Engl. J. Med.355, 983991

    (2006).

    68. Tsao, M. S. et al.Prognostic and predictive

    importance of p53 and RAS for adjuvant

    chemotherapy in non small-cell lung cancer.J. Clin.

    Oncol.25, 52405247 (2007).69. Jiang, H. & Yang, L. Y. Cell cycle checkpoint abrogator

    UCN-01 inhibits DNA repair: association with

    attenuation of the interaction of XPA and ERCC1

    nucleotide excision repair proteins. Cancer Res.59,

    45294534 (1999).

    70. Matthews, D. J. et al.Pharmacological abrogation of

    S-phase checkpoint enhances the anti-tumor activity

    of gemcitabine in vivo. Cell Cycle6, 104110 (2007).71. Syljuasen, R. G. et al.Inhibition of human Chk1

    causes increased initiation of DNA replication,

    phosphorylation of ATR targets, and DNA breakage.

    Mol. Cell Biol.25, 35533562 (2005).

    72. Hartley, K. O. et al.DNA-dependent protein kinase

    catalytic subunit: a relative of phosphatidylinositol 3-kinase and the ataxia telangiectasia gene product.

    Cell82, 849856 (1995).

    73. Blunt, T. et al.Defective DNA-dependent protein

    kinase activity is linked to VDJ recombination and

    DNA repair defects associated with the murine scid

    mutation. Cell80, 813823 (1995).

    This paper identifies DNAPK and shows that it can

    be used as a target to increase toxicity following

    ionizing radiation.

    74. Monfar, M. et al.Activation of pp70/85 S6 kinases

    in interleukin-2-responsive lymphoid cells is

    mediated by phosphatidylinositol 3-kinase and

    inhibited by cyclic AMP. Mol. Cell Biol.15, 326337

    (1995).

    75. Wipf, P. & Halter, R. J. Chemistry and biology of

    wortmannin. Org. Biomol. Chem.3, 20532061

    (2005).

    76. Leahy, J. J. et al.Identification of a highly potent and

    selective DNA-dependent protein kinase (DNA-PK)

    inhibitor (NU7441) by screening of chromenone

    libraries. Bioorg. Med. Chem. Lett.14, 60836087

    (2004).

    77. Zhao, Y. et al.Preclinical evaluation of a potent novel

    DNA-dependent protein kinase inhibitor NU7441.

    Cancer Res.66, 53545362 (2006).78. Farmer, H. et al.Targeting the DNA repair defect in

    BRCA mutant cells as a therapeutic strategy. Nature

    434, 917921 (2005).

    79. Bryant, H. E. et al.Specific killing of BRCA2-deficient

    tumours with inhibitors of poly(ADP-ribose)polymerase.

    Nature434, 913917 (2005).

    References 78 and 79 show that PARP inhibitors

    can be used to selectively kill BRCA1- or BRCA2-

    defective tumours.

    80. Moynahan, M. E., Chiu, J. W., Koller, B. H. & Jasin, M.

    Brca1 controls homology-directed DNA repair. Mol.

    Cell4, 511518 (1999).

    81. Moynahan, M. E., Pierce, A. J. & Jasin, M. BRCA2 is

    required for homology-directed repair of chromosomal

    breaks. Mol. Cell7, 263272 (2001).

    82. Patel, K. J. et al.Involvement of Brca2 in DNA repair.Mol. Cell1, 347357 (1998).

    83. Lomonosov,