• Keine Ergebnisse gefunden

How to kill tumor cells with inhibitors of poly(ADP-ribosyl)ation

N/A
N/A
Protected

Academic year: 2022

Aktie "How to kill tumor cells with inhibitors of poly(ADP-ribosyl)ation"

Copied!
15
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

How to kill tumor cells with inhibitors of poly(ADP-ribosyl)ation

Aswin Mangerich and Alexander BUrkle

Molecular Toxicology Group, University of Konstanz, Konstanz, Germany

ee ,

Poly(ADP-ribosyl)ation is a po$t,translational modification catalyzed by the en~me family of poly(ADP-ribose) polymerases (PARPs). PARPs exhibit pleiot(opic; cellular functions ranging from maintenance of genomic stabUityand chromatin remodeling to regulation of eel! death, thereby rendering PARP homologues promising tar~ets In cancertherapy.Depending on the molecular status of.~ ca:ocercett,low~molecutar weight PARPe inhibitors can (t)eltherbe used'asmonotherapeutic agents followingtheconcept~f synthetic lethality or (11) to support classical chemotherapy orradiothEi~apy.The rationales are the foUowing:(t)in cancers with selective defects in homologous recombination repair, inactivation of PARPs directly causes cell death. In cancer treatment, this phenomenon can be employedtospecificaUy target tlimor cells while sparing nonmalignant tissue. (ii)PARP inhibitorsClln also be used to sensitize cells t!lcytotoxic DNA-damaging treatments. as some PARPs actively participate ingenornicmaii'itenance. Apart from that, PARPhihibietors possess antiangiogenlcfuoctions, thuse opening up a further opti?llto irihibit tUrn?t growth. In view eof the above, a number of high-potencyPARP.inhibitors have been developed during thetast deecadeandare currently evaluated as cancer therapeutics in cUnical trials bysllveral teading pharmaceutical companies.

The Role of Poly(ADP-rlbosyl)ation In Carcinogenesis Carcinogenesis is a multistep process, in which genetic altera- tions drive the transformation of normal cells into malignant ones, ultimately causing cancer. Acquired mutations in onco- genes leading to dominant gain-of-function or tumor sup- pressor genes leading to recessive loss-of-function represent a hallmark of most if not all human cancers,! While oncogene products, such as Ras and c-Myc, largely comprise proteins involved in mitogenic cell signaling leading to increased cel- lular proliferation, tumor suppressor genes, such as DNA repair factors and p53, are mainly involved in the mainte- nance of genomic stability and cell cycle regulation, respec- tively.1 In mammalian cells at least six major and partly over-

Key words: PARP inhibitors, poly(ADP-ribosyl)ation, cancer, synthetic lethality, chemotherapy, radiotherapy, angiogenesis Abbreviations: BER: base excision repair; BRCA: breast cancer susceptibility gene; DSB: DNA double-strand break; HR:

homologous recombination; IR: ionizing radiation; MGMT:

G6-methyl guanine methyltransferase; MMR: mismatch repair;

NHEJ: nonhomologous end joining; PAR: poly(ADP-ribose); PARP:

poly(ADP-ribose) polymerase; PARPi: PARP inhibition; SSB: DNA single-strand break; TMZ: temozolomide, TNKS: tankyrase 001: 1O.IO02/ijc.25683

Correspondence to: Alexander Blirkle, Chair of Molecular Toxicology, Department of Biology, University of Konstanz, D-78457 Konstanz, Germany. E-mail: alexander.buerkle@

uni-konstanz.de

lapping DNA repair mechanisms exist: 06-methyl guanine methyltransferase (MGMT), base excision repair (BER), nu- cleotide excision repair (NER), mismatch repair (MMR) and DNA double-strand break (DSB) repair including the sub- pathways homologous recombination (HR) and nonhomolo- gous end joining (NHEJ).2 Because of the action of turn or suppressor mechanisms such as DNA repair and cell cycle control, acquired mutations are usually rare. However, if tu- mor suppressor mechanisms fail this can cause high mutation rates and sustained genomic instability, both of which are known to drive the evolution of cancers. On the other hand, specific DNA repair mechanisms still need to be functional within tumor cells to ensure cellular survival. I Moreover, the functionality of specific DNA repair pathways represents an important mechanism of cancer resistance to DNA-damaging chemo- and radiotherapy. Thus, paradOxically, targeting tu- mor suppressor mechanisms such as DNA repair is an attrac- tive approach in cancer therapy to sensitize tumor cells to cy- totoxic treatments and to overcome acquired resistance to these treatments.3 Consequently, several DNA repair inhibi- tors are currently being developed as anticancer drugs, and among the most advanced are inhibitors of poly(ADP- ribosyl)ation.

Poly(ADP-ribosyl)ation is a post-translational modifica- tion catalyzed by enzymes of the family of poly(ADP-ribose) polymerases (PARPs). PARPs use NADT as a substrate to form the linear or branched biopolymer poly(ADP-ribose) (PAR) and the by-product nicotinamide (Fig. 1).4 While PARPs can covalently modify themselves (automodification) or other proteins (heteromodification), PAR can also bind noncovalently to numerous proteins via at least three

Konstanzer Online-Publikations-System (KOPS) URL: http://nbn-resolving.de/urn:nbn:de:bsz:352-135413 The definitive version is available at www3.interscience.wiley.com

(2)

• DNA repair/genomic maintenance

• Chromatin remodeling

• Epigenetic regulation

• Regulation of transcription

• Cell cycle regulation

• Regulation of cell death (apoptosis/necrosis)

Figure 1. Poly(ADP·ribose) structure PARPs cleave the glycosidic bond of NAD' between nicotinamide and ribose followed by the covalent modification of acceptor proteins with an ADP·ribosyl unit. PARPs also catalyze an adduct elongation, giving rise to linear polymers with chain lengths of up to 200 ADP·ribosyl units, characterized by their unique ribose (1"->2') ribose phosphate-phosphate backbone. At least some of the PARP family members also catalyze a branching reaction by creating ribose (1'" -> 2'1 ribose linkages. The sites of hydrolysis catalyzed by poly(ADP·ribose) glycohydrolase (PARG) are indicated by arrows. So far, PARP activity has been unequivocally identified for PARP1, PARP2, PARP4 and tankyrases 1 and 2. PAR formation is implicated in a multitude of cellular functions many of which are related to carcinogenesis and development oftumor resistance.

Reprinted with modifications from Ref. 4, with permission from Blackwell Pub. on behalf of the Federation of European Biochemical Societies.

(3)

different PAR binding motifs.5,6 Such PAR-binding motifs include (i) a conserved 20 amino acid motiC (ii) distinct macrodomains8-1O and (iii) a PAR-binding zinc finger, !l,ll all of which fulfill diverse functions in DNA damage response and chromatin remodeling. Importantly, the formation of PAR is transient, since the polymer is degraded by the cata- lytic counterpart of PARPs, i.e., poly(ADP-ribose) glycohy- drolase (P ARG). 13

In humans, 17 homologous genes comprising a conserved PARP signature motif have been identified. However, some of them seem to be catalytically inactive or possess mono- (ADP-ribosyl)ation rather than poly(ADP-ribosyl)ation activ- ity. l4.l5 Several members of the PARP family have been implicated in carcinogenesis as described below.

PARP1 is the founding member of the family of PARPs with pleiotropic function in DNA repair,6 gene transcription,l6 epigenetic regulationl7 and the regulation of cell death and inflammation.18 PARPI accounts for 75-90% of cellular PAR formation upon genotoxic stress, as induced, for instance, by ionizing radiation (IR) or alkylating agents. PARP1 is directly activated by DNA DSBs and single-strand breaks (SSBs) as well as by some unusual DNA structures.4 Apart from that PARPI can be activated by mitogenic stimuli through the ERK signaling pathway.19 Upon DNA damage, PAR formation leads to the recruitment of several DNA repair factors, such as XRCCI and DNA ligase III, to the site of the damage. In addi- tion, poly(ADP-ribosyl)ation of these factors is able to regulate their activity status. Altogether, this is believed to activate and facilitate DNA repair signaling.6 Specifically, PARP1 partici- pates in BER and an alternative pathway ofNHEJ. Moreover, a role in NER and the regulation of HR was reported.20-22

Pur- thermore, the MMR proteins MSH2 and MSH6 bind PAR in a noncovalent manner. s PARP1 also functions as a regulator of telomere length2324 and is involved in organizing the spindle apparatus.2526

Importantly, PARP1 also regulates the nuclear localization and activity of the tumor suppressor gene pS3.27,28 In view of the above, PARP1 is considered a general caretaker of the genome.29 Accordingly, P ARP I-deficient mice and cells are hypersensitive to IR and alkylating agents.30 The fact that PARP1-deficient mice are prone to induced tumor formation and show an increased age-dependent spontaneous tumor load suggests a role for PARP1 as a tumor suppressor gene.31 ,32 In this regard, it is interesting to mention that the formation of some human cancers has been linked to a hypomorphic single nucleotide polymorphism in the gene encoding PARPI, lead- ing to the amino acid replacement V762A.33 Moreover, PARP1 expression is upregulated in various types of human cancers, and this has been linked to resistance of cancer cells to genotoxic treatment.,l3-.l7 In stark contrast, PARPl activity was also discussed as a tumor promoter in a mouse model of skin carcinogenesis, which may be related to its function as an inducer of cell death and tissue inflammation in specific condi- tions.38 We recently generated a mouse model with ectopic expression of human P ARP-l, which lead to premature devel- opment of inflammation-related chronic diseases and further

supported the view of a complex and double-edged role of PARP-1 during carcinogenesis.39,4o Thus, the age-related tu- mor-spectrum of those mice was shifted showing lower inci- dence of sarcomas, but increased incidence of carcinomas, which may be related to the development of inflammatory diseases.

Apart from P ARP I, P ARP2 is the only other P ARP family member known to be activated by binding to DNA struc- tures. It accounts for most of the residual nuclear PAR for- mation upon DNA damage and physically and functionally interacts with PARPl. PARPI and PARP2 exhibit, at least in part, redundant functions. This is supported by partially overlapping phenotypes of the corresponding knock-out mice and by the fact that double deficiency results in embryonic lethality in the mouse.41,42 Recently, functions of PARP2 in- dependent of PARPl, such as in genomic maintenance, gene transcription, and T-cell development, were reported.41 Although PARP2-deficient mice do not show enhanced spon- taneous tumor development, there is rapid development of spontaneous T -cell lymphomas in a pS3-deficient back- ground, suggesting that P ARP2 may also act as a tumor sup- pressor gene in specific genetic constellations.43

PARP3 mainly resides in the nucleus;!1 where it mayex- hibit mono(ADP-ribosyl)ation activity.45 During cell division, P ARP3 localizes to centrosomes and is involved in the regu- lation of Gl/S cell cycle progression.46 Although a direct role of PARP3 in DNA repair is still a matter of debate,45 it is striking that the major interaction partners of P ARP3 are involved in the NHEJ pathway. Moreover, PARP3 immuno- precipitates with DNA ligase III and is able to activate PARPI in the absence of DNA, which links it to BER.44

PARP4 also known as Vault PARP (VPARP) is part of the cytoplasmic vault ribonucleoprotein complex, which has been implicated in multidrug resistance. VP ARP has been localized to the nuclear pore and the mitotic spindle and exhibits poly(ADP-ribosyl)ation activity.47 VPARP-deficient mice show an increase in carcinogen-induced colon and lung tumor incidence as well as reduced tumor latency:18

P ARPSa and P ARPSb, better known as tankyrases (TNKS) I and 2, exhibit poly(ADP-ribosyl)ation activity. They localize to multiple subcellular sites including cytoplasmic membrane compartments, telomeres and spindle poles. While TNKS1 was reported to act as a positive regulator of telomere length and is required to resolve sister telomeres during mitosis, the role of TNKS2 in telomere length regulation remains to be determined.49 Apart from its role in telomere regulation, TNKSI was also implicated in GLUT4 vesicle trafficking:19 Both tankyrases seem to exhibit at least in part redundant functions, since Tnksl and Tnks2 single knock-out mice are viable, whereas double deficiency is embryonically letha1.50 Expression analyses showed that tankyrase expression levels are upregulated in a variety of human cancers, indicating a direct role of tankyrases in carcinogenesis.49

PARPIO is localized in the nucleus and the cytoplasm and was reported to possess mono(ADP-ribosyl)ation activity. It

(4)

Synthetic lethality

HR.deficicmt cells:

Genetically or through Inhibitor treatment

Chemo-and Radiosensltization

Induction of DNA damagethrO\.lgh DNA damaging agents and

IR

Inhibition of angiogenesis

Tumor-driven angiogenesis

Inhibition of poly(ADP-ribosyl)ation

Induction oftumor cell death and senescence

Figure 2. Overview of how PARP inhibition can support cancer therapy. DSBs indicate DNA double-strand breaks; HR: homologous recombination; IR: ionizing radiation; SSBs: DNA single-strand breaks_

was identified as a c-MYC interacting protein, suppressing cellular transformation induced by c-MYC and E1A protein.51,52

PARP9, PARP14 and PARP15 belong to the subfamily of macro- P ARPs. While P ARP9 is catalytically inactive, P ARP 14 possesses mono(ADP-ribosyl)ation activity and PARP15 ei- ther mono- or poly(ADP-ribosyl)ation activity.52,53 They are localized mainly in the nucleus and contain macrodomains.

In general, macrodomains are highly conserved structures that are often involved in the setting of the chromatin status.

Macro-PARPs in particular are involved in the transcriptional regulation in response to cytokines and have been linked to carcinogenesis.54 Because of their differential expression in B- cell lymphomas, they were also designated as B aggressive lymphoma (BAL) proteins. P ARP9 was originally identified in a genome-wide search for risk-related genes in diffuse large B-cell lymphoma and its expression was significantly increased in chemoresistant tumors.53

In summary, several P ARP family members play active roles in carcinogenesis, which renders them interesting drug targets for cancer therapy. Such an approach gains even fur- ther attraction by the findings that at least short term ab- sence of P ARP function seems to have only minor physiolog- ical consequences in the absence of DNA damage and genomic instability. Interestingly, in a preclinical mouse model also long-term treatment with a highly potent PARP inhibitor (AZD2281) did not result in any obvious signs of toxicity.55 Consequently, various phase I and II clinical trials

are currently underway to evaluate the use of P ARP inhibi- tors in treatment of a variety of tumors including breast can- cers, ovarian cancers, prostate cancer, colorectal cancer, gas- tric cancer, cancer of the small intestine, hepatocellular carcinomas (HCCs), sarcomas, lymphomas, melanomas, pan- creatic neoplasms and glioblastomas. At present one inhibitor, i.e., BSI-201 (Sanofi-Aventis), reached phase III clinical trials for breast cancer and squamous cell lung cancer therapy in combination with gemcitabine/carboplatin (http://

c1inicaltrials.gov) .

In the paragraphs below, we discuss the underlying molec- ular mechanisms of how P ARP inhibition (P ARPi) mediates tumor cell death (Fig. 2). Review articles with a more clinical focus of PARPi have been published recently.50.57

Chemistry and Pharmacology of PARP Inhibitors Most P ARP inhibitors are competitive with the substrate NAD+ and thus interfere with its binding to the enzyme's active site, which is highly conserved between different P ARP homologues. Interestingly, nicotinamide itself, i.e., the by- product of PAR formation, acts as a natural weak inhibitor of PARP activity. Molecular modeling and X-ray crystallogra- phy revealed that the amide of the nicotinamide forms three crucial hydrogen bonds with PARP1 at the hydroxyl group of Ser904 and the amide backbone of Gly863. Furthermore, the pyridyl ring interacts via a 1t-1t stacking with Tyr907.58,59

Based on a good understanding of the structure-activity

(5)

relationship, a significant increase in potency of PARP inhibi- tors was achieved during the last decades. Likewise, their bio- availability was considerably improved.60 Based on their chemical structure, the following classes of P ARP inhibitors can be distinguished:

1. Benzamide analogs: these include nicotinamide and 3- aminobenzamide (3-AB). IC50s of those first generation inhibitors are in the micromolar range. For clinical use those inhibitors exhibit many disadvantages such as low potency, short cellular residence time, nonspecific inter- actions such as the inhibition of mono(ADP-ribosyl)a- tion reactions, and intrinsic antioxidant function.

Whereas benzamide analog P ARP inhibitors mainly have remained experimental tools for mechanistic research,60 there are two interesting exceptions. The first exception is nicotinamide, the by-product of poly(ADP- ribosyl)ation itself, which was tested in clinical trials in combination with radiotherapy;61 the second one is BSI- 201 (Sanofi-Aventis). BSI-201 (4-iodo-3-nitrobenza- mide) is a recently developed compound that inhibits PARP1 and possibly also other enzymes via an irreversi- ble covalent modification. This is in contrast to other common P ARP inhibitors and may explain the excep- tional efficacy of BSI -201 in several clinical trials.62 2. Isoquinolinones and dihydroisoquinolinones: these

group of compounds, which include the inhibitors 5- AIQ, DPQ and GPI-6150, were reported as consider- ably more potent than 3-AB but were mainly used as experimental tools as well. Their IC50s are in the upper nanomolar range.60

3. Benzimidazoles, indoles and related compounds include the inhibitors NU 1085, ABT-888 (Abbott) and AG14447 (AGOI4699, phosphate salt of AG14447;

pfizer) with IC50s in the low nanomolar range.60,63.64 ABT -888 is currently in phase I clinical trials as single- agent and combination treatment for solid tumors, leu- kemia and BRCA-deficient tumors. It is also in phase II trials for the treatment of glioblastoma multiforme in combination with telozolomide (see below). AG014699 entered phase II clinical trials for the treatment of BRCA-deficient tumors.56

4. Phthalazin-1(2H)-ones and quinazolinones comprise the inhibitors NU1025 and AZD2281 (Olaparib; Astra- Zeneca). The most potent of those inhibitors possess IC50s in the low nanomolar range. Pharmacokinetics and pharmacodynamics of olaparib were studied in a phase I clinical trial indicating rapid absorption and elimination and effective in vivo P ARP inhibition. Cur- rently, Olaparib is in phase II clinical trials against vari- ous types of malignancies.56,65

5. Isoindolinones include the potent inhibitor EB-47 with an IC50 value below 100 nM.60

6. Phenanthridinones comprise the inhibitors PJ34 and Ino-1001 (Inotek), the latter with an IC50 value below

100 nM. The inhibitor Ino-1001 was tested in combina- tion therapy with temozolomide in a phase I clinical trial in melanoma patients.60,66

In principle, there are two ration ales for the usage of PARP inhibitors in cancer therapy: (i) use as stand-alone drugs for tumors with a specific genetic make-up, following the "synthetic lethality" concept and (ii) use in combination with DNA-damaging treatments such as chemo- and radiotherapy.

PARP Inhibitors in Monotherapy

Tumor-dependent DNA repair deficiencies represent an attractive target in cancer therapy by following the concept of synthetic lethality.67 Two genes are synthetically lethal if loss of function of either gene alone is compatible with cell sur- vival, but mutation or inactivation of both cause cell death.

As a result, targeting a gene that is synthetic lethal to a can- cer-relevant mutation should kill only malignant cells and spare normal cells.GB The first proof-of-principle of this novel approach in a clinical setting was achieved with PARP inhibi- tors in BRCA-deficient mammary carcinoma cells. The tumor suppressors BRCA1 and BRCA2 participate in HR and mi- totic control.69 Germline mutations in one of the BRCAl or the BRCA2 alleles predispose to the development of several cancers, including breast, ovarian and prostate cancer. Malig- nant cells in patients carrying a heterozygous loss-of-function BRCA mutation typically display loss of heterozygosity. This results in HR deficiency, which causes genetic instability and drives carcinogenesis.70 The loss of HR repair is not shared with normal cells and can be exploited by P ARPi to induce selective tumor cytotoxicity. Mechanistically, cells with inhib- ited P ARP activity may acquire more unrepaired SSBs and stalled or collapsed replication forks caused by endogenous DNA-damaging events. Replication forks contain several pro- teins such as helicases and polymerases, forming the so-called replisome. Usually progression of the replication fork contin- ues until it encounters a replication fork barrier such as DNA-protein complexes or SSBs. In this case the replicative helicase progresses much more slowly, so that the fork is

"stalled." If this goes along with the disassembly of the repli- some the fork "collapses" and a DSBs is formed. 71 Usually DSBs ariSing from collapsed replication forks are repaired by RAD51-dependent HR; however, in cells with loss of function of BRCA1 or BRCA2, HR is defective and repair of these breaks by alternative error-prone DSB repair mechanisms would cause genomic instability, leading to cell death. Over- all, there is ample evidence supporting such a model with regard to PARPi: Loss of PARP1 function can induce the for- mation of nuclear RAD51 foci as well as sister chromatid exchange in BRCA-proficient cells, indicating the repair of DSBs. In contrast, no RAD51 foci were formed in BRCA-de- ficient cells. Instead, PARP inhibitor-treated BRCA-deficient cells showed more chromatid aberrations compared to wt

(6)

cells. Furthermore, PARPi induced the formation of y-H2A.x foci, which indicate sites of DNA damage.7273 Such a pheno- type suggests failure of DSB repair by conservative RADS1- dependent sister-chromatid recombination and activation of alternative error-prone pathways such as NHEJ.72.74

Interest- ingly, PARP1 itself participates in the reactivation of stalled replication forks.7s It binds to and is activated at stalled repli- cation forks and mediates the recruitment of Mrell to pro- mote replication fork restart after release from replication blocks.76,77 On the other hand, PARPi could also have an effect on the repair of occurring DSBs by alternative mecha- nisms, e.g., via its involvement in NHEJ.78 RNAi experiments supported the view that PARP1 rather than PARP2 is re- sponsible for the repair of toxic recombinogenic lesions that spontaneously occur in human cells.n.73 Thus, depletion of P ARP2 and BRCA2 had no effect on clonogenic survival of the cells, and depletion of PARP2 in PARPl and BRCA-2- depleted cells did not result in additional toxicity.

The concept of synthetic lethality represents one of the few examples, where the mutation that caused carcinogenesis can be exploited for targeted cancer therapy. Recently, a phase I clinical trial with Olaparib in patients enriched in carriers of BRCA mutations was conducted?9 P ARPi was achieved successfully in peripheral blood lymphocytes, hair follicle cells and tumor tissue. Carriers of BRCA mutations did not show an increased risk of adverse effects. In line with the cell culture data mentioned above, P ARPi in patients' hair follicle cells was rapidly associated with downstream induction of DSBs.79 Although most patients with BRCA mutations showed a response in this and another phase II study, not all BR CA mutation carrying patients did SO.79,80

The authors hypothesized that this may have resulted from preexisting genetic resistance; thus, it was shown that second- ary BRCA2 mutations may restore BRCA function and HR, thereby causing resistance to PARPi.SI,82 Moreover, the effec- tiveness of PARP-inhibitor-dependent cell death in BRCA deficiency depended on the use of highly potent P ARP inhib- itors.73,83,84 Although the inhibitors used in the studies described here were reported to exhibit selectivity for PARP1 and PARP2 with 1-3 orders of magnitude over PARP3, vPARP and tankyrase,73 specificities for other PARPs were not tested. Therefore, the specificity of the synthetic lethal mechanism with regard to PARP1 inhibition is not entirely clear. In this regard, it is interesting to mention that synthetic lethal interactions also exist in cells deficient for both TNKS1 and BRCA.85

In future studies, it will be interesting to expand this con- cept to cancers with other defects in HR, such as loss of function of RADS1, ATM, ATR and CHKl and CHK2, as well as components of the Fanconi's anemia repair pathway.

First preclinical data already showed promising results. Thus, ATM-deficient cells were shown to be sensitive to PARPi.86 Moreover, P ARPi sensitizes A TM -deficient cells to DNA- damaging agents?O These data are consistent with the finding that PARPi leads to the activation of ATM, which then indu-

ces the subsequent HR repair.86 Mutations in the A TM gene were detected in T-cell leukemia, mantle-cell lymphoma and chronic B-lymphocytic leukemia, rendering tumors with such mutations ideal candidates for PARPi therapy.s6 In addition, deficiency of the PTEN tumor suppressor gene causes a HR defect. Consequently, PTEN deficiency sensitized tumor cells to potent PARP inhibitors.s7 Viewed together, there is a con- siderable potential to extend monotherapy with PARP inhibi- tors to a larger group of patients with general defects in HR.88,89 Thus, tumors showing "BRCAness" may represent another promising target for P ARPi. For instance, some spo- radic breast or ovarian cancers show epigenetic inactivation of BRCAl.9o

PARP Inhibitors in Combination Therapy

Many cancer therapies act by causing DNA damage, which, if unrepaired, triggers cell death. Consequently, DNA repair mechanisms may render cancer cells resistant to chemother- apy or radiotherapy. Several PARPs, especially PARP1 and PARP2, actively participate in several DNA repair mecha- nisms, in particular in the repair of SSBs. Therefore, P ARPi sensitizes tumor cells to cytotoxic DNA-damaging agents and IR, thereby improving the therapeutic index of those thera- pies. Consequently, several preclinical and clinical studies using P ARP inhibitors in combination with cytotoxic agents including monofunctional alkylating agents, such as temozo- lomide, topoisomerase-I poisons, such as topotecan, and DNA-crosslinking agents, such as cisplatin, have been con- ducted. The data showed that P ARPi sensitizes malignant cells to all of these agents (summarized in Table 1). Likewise, P ARPi sensitizes tumor cells to IR, in vitro and in vivo.

PARP inhibitors as chemosensitizers

The DNA-methylating agent temozolomide (TMZ) is rapidly converted into an active intermediate, i.e., S-(3-methyltria- zen-lyl)imidazol-4-carboxamide. TMZ can cross the blood brain barrier efficiently and exhibits limited bone marrow toxicity. TMZ is used as a stand-alone drug or in combina- tion with IR in the therapy of central nervous system malig- nancies, such as glioblastoma multiforme and melanomas.

TMZ acts via the addition of methyl adducts to three main positions in nucleobases, i.e., N7-methylguanine (~70% of adducts), 06-methylguanine (~S% of adducts) and N3-meth- yladenine (~9% of adducts).91 The mechanism of action of TMZ-induced cytotoxicity is mainly through the formation of 06-methylguanine. Thus, during replication 06-methylgua- nine incorrectly pairs with thymine, leading to the induction of MMR. However, the repair of the mismatched base mainly leads to the reinsertion of thymine, resulting in futile cycles of repetitive DNA repair, finally causing DSB formation and apoptosis.91 Resistance to TMZ is frequently observed by two different mechanisms: (i) Upregulation of MGMT activity and subsequent efficient repair of 06- methylguanine.92 (ii) Development of MMR deficiency: In the absence of MMR the cell is able to survive despite the presence of 06_

(7)

Table 1. PARP inhibitors as chemo- and radiosensitizers Combination

. agent Temozolomide

Platinum druis

Topoisomerase 1 inhibitors

Mode of action w/o PARPi

" Alkylating agent

o Formation of N7methylguanine, 06-methylguanine, N3-methyladenine

" Cytotoxicity mainly elicited via the formation of 06-methylguanine by induction of futile cycles of MMR and subsequent formation of DSBs

" Development of tumor resistance by upregulation of MGMT and devel- opment of MMR deficiency

• Terminal phenotype: mainly apoptosis and necrosis

• Introduction of inter'c;lnd.

intrastrand crosslinks

o Can be repaired by NER

• Terminal phenotype: mainly apoptosisand necrosis

" Introduction of transient SSBs by Topol

• Topol inhibitors prevent religation of DNA strand breaks causing persistent SSBs and stalled replication forks

" Formation of DSBs during replication

• Terminal phenotype: mainly apoptosis and necrosis

• Introduction of SSBs ahdDSBS (",25:1) . . ..•...

• SSBs maybecomelethaldWing replication, when.DSBsar~formed by replication forK collapse

" Termlr:ralphenotype: mainly apoEt?~i:~n.~ ~~5r?_Si$

Mode of action w/PARPi

• PARPi restrains BER

" Other adducts such as N-methylpurines are not repaired efficiently

" Accumulation of SSBs

" Conversion of SSBs to DSBs during replication

• Terminal phenotype:

mainly apoptosis

• PARPi interferes with NER

"Inhibition of PARP·1, which binds to platinum compound~

induced damage and may be directly involved in its repair

• Terminal phenotype: mainly apoptosis

" PARPi prevents repair of SSBs introduced by Topol inhibition

• PARPi counteracts Topol activity, since PARP-l induces the activity of Topol

" Conversion of SSBs to DSBs during replication

" PARPi prevents repair of SSBs

• ·ConversionofSSBs to DSB.s during replication

. • Terminal phenotype:apoptosis,

aut9phagyandsene.s.~ence

Current clinical status

• Clinical trials for the treatment of:

o metastatic melanoma (phase I) o metastatic prostate cancer (phase I)

o colorectal cancer (phase 11)

o breast cancer (phase 11) o glioblastoma multiforme (phase 11)

o childhood neuroblastoma

o solid tumors (phase I) o leukemia (phase I)

" Clinical trials forthe tr!!atment of:

bre.ast cancer (phase m) o ovarian cancer (phase 11)

ci advanced solid tumors . (phase!)

b sql,lamous cell lung cancer (phase Ill)

" Clinical trials for the treatment of:

o ovarian cancer (phase 11) o colorectal cancer (phase I)

o advanced solid tumors (phase I)

o lymphoma (phase I)

.... CHnicaitrials forthe treatment of:

o.c.N$neoPlas!lis(phase I)

For references, see text. Clinical trials according to http://ciinicaltrials.gov. BER indicates base excision repair; DSBs: DNA double-strand breaks; IR:

ionizing radiation; NER: nucleotide excision repair; SSBs: DNA single-strand breaks; Topol, topoisomerase 1.

methylguanine, since MMR is required for the induction of DNA-strand breaks after the formation of 06- methylguanine.

P ARP activity in patients' lymphocytes is increased at 4h af- ter TMZ administration due to DNA damage induction. As P ARPi restrains BER, other lesions of methylpurines which are usually efficiently repaired by BER become lethal via the induction of apoptosis.91 Consequently, PARPi following TMZ treatment resulted in increased peak levels of DNA- strand breaks and the persistence of N-methylpurines, most likely increasing cytotoxicity.93 Presumably, potentiation of TMZ cytotoxicity by P ARPi requires the conversion of SSBs to DSBs in actively dividing cells passing S-phase, as shown recently in an analogous cell culture model using P ARPi in combination with the DNA methylating agent methyl metha- nesulfonate (MMS).94 Therefore, in general, a PARP inhibitor that has a long half-life or can accumulate for a long period of time in tumors is preferred for optimal potentiation of TMZ.95 Results of several studies suggested that PARPi

restores TMZ sensitivity and increases its efficacy in particu- larly in MMR-deficient tumors.96-98 However, the antitumor effect of TMZ was also modestly enhanced by P ARPi in MMR-proficient cells9698

perhaps due to additive effects resulting from increased number of cytotoxic lesions. Thus, under conditions of P ARPi, N7 -guanine and N3 -adenine adducts may contribute to additional cell death.96 Based on these data, future prescreening of cancer patients for the MMR status of tumor cells may help to predict the outcome of the combination therapy.96 With regards to the questions which P ARP homologues are responsible for the sensitization effect, PARPl is most likely the main contributor to TMZ sensitization, as PARPl-silenced melanoma cells were extremely sensitive to TMZ.99 However, it was shown recently that both PARPl-proficient as well as PARPl- silenced cells show increased sensitivity to TMZ/P ARPi com- bination treatment.IOO The latter study suggests that P ARP2 or other P ARPs are also involved in the repair of DNA

(8)

damage introduced by TMZ. In summary, P ARPi has been shown to potentiate antitumor effects of TMZ in the treat- ment ofleukemia,lol glioma,96.102.103 lung64104 and colon car- cinoma, both in vitro64.97.I03 and in xenograft preclinical models.104105 Consequently, PARP inhibitors are currently being tested in clinical combination therapy with TMZ for the treatment of metastatic malignant melanoma,66.<J2,I06 glio- blastoma multiforme,107 childhood neuroblastoma, 108 advanced solid tumors,109 refractory solid tumors and Iymphomas. llo

Platinum drugs induce inter- and intrastrand crosslinks that are removed by NER. Because PARP! participates in NER, synergy between P ARP inhibitors and platinum drugs is expected?I.22 Moreover, PARP-l was reported to bind to DNA damages induced by platinum compounds, suggesting a direct role of P ARP-l in the repair of such damages.I l l112 Accordingly, several studies suggest that P ARPi potentiates the effect of platinum compounds. Thus, it was shown that P ARPi increases the cytotoxicity of platinum complexes in cisplatin-resistant ovarian tumor cells. 11 3 Another study showed selective synergy between P ARPi and cisplatin in BRCA2-deficient mammary tumor cells.l14 Moreover, combi- nation of PARPi (AZD2281) with platinum compounds increased the survival in a genetically engineered mouse model of hereditary breast cancer.S5 Nevertheless, most tumors in this study could not be eradicated with P ARPi/

platinum combination schedules indicating the existence or development of resistant tumor cells.s5 Also in another study cotreatment of spontaneously occurring tumors with P ARPi (AZD2281) and carboplatin in a tissue-specific BRCA and pS3-deficient mouse model showed no advantage over carbo- platin monotherapy. However, if PARPi was continued as a monotherapy, the time to tumor relapse or death was signifi- cantly increased, indicating that such a therapy schedule rep- resents a beneficial treatment option. llS The latter study also suggested that effectiveness of P ARPi and platinum com- pounds are both dependent on functional HR indicating that the mechanism of platinum-induced DNA damage response is not fully understood yet. Contrasting reports about phar- macotoxicology of platinum/P ARPi cotreatment exist in the literature: while increased nephrotoxicity of cisplatin in com- bination with PARPi (AZD2281) was observed in one study,S5 cotreatment of cisplatin with P ARP inhibitors (3AB, BGP-lS) was reported to attenuate cisplatin-induced nephro- toxicity in other studies.116.117 Consistent with the notion that activation of P ARP is implicated in the etiology of cis- platin-induced toxicity, PAR synthesis was found to be enhanced upon cisplatin treatment in renal proximal tubular cells. Jl7 This aspect calls for further research, as it represents a potential mechanism of lowering cisplatin-induced toxicity.

Currently, phase I and II clinical trials are conducted using PARP inhibitors in combination with platinum compounds against "triple negative" and BRCA-mutated breast cancers, ovarian cancers as well as advanced solid tumors.56 More- over, the PARP inhibitor BSI-20l (Sanofi-Aventis) is in phase

III clinical trials for breast cancer and squamous cell lung cancer therapy in combination with gemcitabine/carboplatin.

(http://clinicaltrials.gov).

Nuclear DNA topoisomerase 1 (Topo!) introduces a tran- sient SSB by forming a covalent DNA-Topo! complex, which allows for the rotation of DNA around the intact strand and relaxation of supercoiling. Thereafter, religation of the SSB restores the DNA integrity and the enzyme diSSOCiates from the DNA.lI8 Topo! is inhibited by the alkaloid camptothecin, which is the parent compound of the anticancer agents irino- tecan and topotecan, currently used to treat ovarian, cervical and small cell lung cancer. Camptothecins induce formation of a stable DNA-Topol complex preventing religation of the DNA-strand break and dissociation of the enzyme. I IS This leads to increased numbers of SSBs and stalled replication forks, which require PARP-mediated repair. Although the DNA-Topo! complex does per se not induce cell death, it may become lethal during replication, when DSBs are formed by collapse of stalled replication forks during S-phase.ll9 Of note, PARPl itself directly interacts with and induces the ac- tivity of Topo!. Therefore, combination treatment of Topo!

inhibitors with PARP inhibitors may enhance tumor cell cy- totoxicity by counteracting Topol activity and favoring the accumulation of SSBs and unresolved replication forks.120-122 Consequently, PARPi in combination with topotecan treat- ment is effective in different cell lines of lung, colon, ovary and breast cancers as well as in human neuroblastoma and colon adenocarcinoma xenografts.104.108.123 Currently, phase I and II clinical trials are conducted with P ARP inhibitors in combination with Topo! inhibitors against ovarian cancer, advanced colorectal cancer and other advanced solid tumors.56

Topoisomerase 2 (Top02) can resolve topological prob- lems in DNA structures by introducing a transient DSB, and it is involved in several nuclear functions, including chromo- some segregation, DNA replication and chromosome organi- zation. Inhibition of Top02 activity and induction of enzyme- mediated DNA damage is an effective strategy for chemo- therapy.124 However, in contrast to sensitization of Topol, data of P ARPi in combination with Top02 inhibitors are less conclusive. P ARPi did not increase cytotoxicity caused by the Top02 inhibitor etoposide. Etoposide did not activate PARP even at concentrations that caused significant levels of apo- ptosis. m Moreover, the classical Top02 inhibitor amsacrine (m-AMSA) affected mouse cells independently of their PARPl status. However, studies on triazoloacridones yielded contrasting results. These compounds were also reported to be inhibitors of Top02 and were shown to exhibit very strong cytotoxic activity toward malignant cells in vitro and a strong cytostatic effect against solid tumors in nude mice. Within this class of inhibitors, the potent Top02 inhibitor C-130S strongly affected proliferation of PARPI-deficient fibroblasts, whereas the action of less active compound C-lS33 toward normal and PARPl-deficient cells was comparable. While C- 1305 had only weak effects on DNA damage and apoptosis,

(9)

it strongly affected cell-cycle progression in normal and PARPI-mutant cells and arrested both cell types in G2-M phase. This G2-M arrest was greatly prolonged in PARPI-de- ficient cells as compared with wild-type fibroblasts. However, other studies showed that metabolites of triazoloacridones co- valently bind to DNA, which may explain at least in part the conflicting results with classical Top02 inhibitors and could provide an alternative mechanism of interference with P ARP function in the case of triazoloacridones usage.126

PARP inhibitors as radiosensitizers

Ionizing radiation mediates cytotoxicity via induction of DNA damage, i.e., SSBs and DSBs at a ratio of~2S:I.127 SSBs are mainly repaired by BER, in which PARPI and P ARP2 exhibit crucial functions as mentioned above. 128 In analogy to camptothecin toxicity, SSBs induced by IR are presumably not cytotoxic per se, but may become lethal dur- ing replication, when DSB are formed by replication fork col- lapse.119 This view is compatible with findings showing that P ARPi increased replication-dependent DSBs and delayed repair of radiation-induced DNA breaks associated with yH2A.x and RADSI foci, which are indicators of HR repair. 129.1 30 Furthermore, radiosensitizing effects of PARPi are pronounced in dividing cells, but abolished in growth- arrested cells.129-131 PARPi potentiated IR-induced cytotoxic- ity in PARPI-proficient, but not in PARPI-deficient cells, indicating that PARPI is the main contributor to IR-insensi- tivity. IJ2 However, P ARPi and ablation produce different out- comes concerning radiosensitivity, i.e., P ARPi radiosensitizes cells specifically in S-phase, whereas silencing of PARPI ren- ders cells hypersensitive to IR regardless of cell cycle status, demonstrating that PARPI has functions either dependent on its enzymatic activity or not.130.133 A further level of com-

pexity is added by the finding that PARPI is an essential mediator of IR-induced NF-kappaB activation, which in turn is an important mediator of resistance to IR. Thus, PARPi sensitized p6S-proficient but not the p6S-deficient cells to IR.

Therefore, potentiation of IR-induced cytotoxicity by a potent PARP inhibitor (AG14361) was thought to be mediated by inhibition of NF-kappaB activation. 134

The observation that P ARPi is equally effective to radio- sensitize cells in normoxic or hypoxic conditions may be clinically relevant, since a hypoxic environment can drive the selection of aggressive turn or cell phenotypes through increased genomic instability and downregulation of DNA repair. Hypoxic cells are about 3-fold more radioresistant than normoxic cells, and intratumoral hypoxia has been described as a significant source of treatment failure using radiotherapy. us

Currently, P ARP inhibitors are evaluated as radiosensi- tizers in phase I and II clinical trials of treatment of head and neck cancers as well as CNS neoplasms. In this regard, it is interesting to note that glioblastoma cells are a rapidly dividing cell population within the nonreplicating tissue of the normal brain and are therefore an attractive target for

P ARPi/IR combination therapy, especially in view of the fact that P ARPi potentiates the cytotoxicity of TMZ, which is of- ten used in combination with radiation therapy.129

PARP Inhibitors as Antiangiogenic Agents

Usually, once a normal tissue is formed in the adult orga- nism, the formation of new blood vessels is a tightly con- trolled mechanism. However, tumors depend on the estab- lishment of microcirculation to grow beyond a few millimeters. Moreover, metastatic cells are only released into the circulation after the tumor established its own microcir- culation. Different sequential steps are necessary for this kind of vasculogenesis. These processes are mediated by angio- genic factors. Therefore, targeting these factors for cancer therapy is currently under investigation.136

A role of PARPI in angiogenesis has been reported previ- ously.137 Thus, PARPi leads to a reduction of tumor vascula- ture and down regulation of the expression of genes involved in angiogenesis during skin carcinogenesis in mice. Differen- ces in tumor-related gene expression, i.e., genes that are involved in inflammation and vasculogenesis, were also found between PARP inhibitor-treated HCC and control xenografts.

Therefore, PARPi appeared to be capable of controlling HCC growth and preventing tumor vasculogenesis by regulating the activation of different genes involved in tumor progres- sion.138 In particular, P ARPi has been demonstrated to reduce the induction of hypoxia-inducible factor-lex (HIF- lex). HIF is a transcription factor that binds to hypoxia- response elements and activates various genes involved in angiogenesis, energy metabolism, cell proliferation and apo- ptosis. 137.139-142 Additionally, PARPI-depleted melanoma xenografts displayed reduced expression of the angiogenesis marker PECAM-I/CD31 and the proinflammatory mediators TNF-Q( and GITR.99 Furthermore, P ARPi at concentrations devoid of cytotoxiC effects abrogated migration in response to vascular endothelial growth factor or placenta growth factor, inhibited formation of tubule-like networks, and impaired angiogenesis in vivo, which can be attributed at least in part to PARPl. 143 Although the role in tumor vasculogenesis is far from understood, the current preclinical results suggest a supportive effect of PARPi in cancer therapy by counteract- ing tumor-neovasculogenesis.

Concluding Remarks

Even though the use of P ARPi in cancer therapy has received much attention in recent years, some issues remain to be addressed carefully in the near future:

An important question is the issue of long-term safety.

The use of P ARP inhibitors was suggested as a prophylactic treatment for individuals with heterozygous BRCA muta- tions.144 However, such proposals should be considered with great caution: A caveat in the systemic long-term treatment with P ARP inhibitors is the impairment of DNA repair and genomic stability in normal cells, which may lead to second- ary tumors at later age. The fact that research on

(10)

homologue-specific P ARP inhibitors is still at an early stage and that Parp1l2 as well as Tnks-1I2 double knock-outs are not viable makes it necessary to carefully balance for risk and benefit of PARPi therapy. Moreover, it needs to be monitored if the unusual mechanism of PARPi by BSI-20l, i.e., via an irreversible, covalent protein modification, affects long-term toxicity during clinical usage differently compared to classical competitive P ARP inhibitors. Another caveat that should be considered is the genetic status of a tumor, as P ARPi can attenuate the cytotoxic effects of cisplatin against RB-deficient tumors. Mechanistically, this outcome was explained by the finding that usually DNA damage-induced death of RB-defi- cient cells was primarily necrotic which is dependent on PARP overactivation. [45 However, due to the promising pre- clinical and clinical data of P ARP inhibitor usage in mono- or combination therapy, the P ARP inhibitors treatment regi- mens against manifest cancer appear justifiable at the current stage.

The route of cell death that is pursued upon P ARPi is another field of research that awaits further clarification: Usu- ally, DNA-damaging agents and IR mediate their lethal effects via enhanced apoptosis, necrosis, mitotic catastrophe and tumor cell senescence.146 Additional P ARPi seems to switch the route of cell death from necrosis to apoptosis, because DNA damage-induced necrosis often depends on P ARP activity. [13.147 If such a switch in the terminal pheno- type of tumor cells has also consequences on the effectiveness of the eradication of the tumors, e.g., enhanced stimulation of tumor-killing immune cells, needs to be determined in future studies. Moreover, enhanced autophagy and senes- cence have been described recently as alternative terminal phenotypes of tumor cells upon P ARPi in combination with radiotherapy.146.148 It will be interesting to see if these termi- nal phenotypes also play a significant role in other treatment regimens.

A further issue that has been analyzed only sporadically includes PARP-inhibitor-dependent off-target effects. Thus, theoretically, P ARP inhibitors may also interfere with

References

NAD+-dependent enzymes other than PARPs, such as mono-ADP-ribosyltransferases or sirtuins and may result in considerable off-target effects.6o Only a few PARP inhibitors, including ABT-888, have been tested for off-target effects on sirtuins and have been shown to be inactive.63

Although genetic and RNAi approaches suggest that sensi- tization and cytotoxic effects of P ARPi depend predominantly on PARPl, the question if other PARP homologues also con- tribute to the antitumor effects is largely unsolved. Therefore, development of homologue-specific P ARP inhibitors is another priority for future research. Progress in this field may also provide the basis for further improvement in P ARPi treatment in order to reduce off-target effects; however, as mentioned before the field of homologue-specific P ARPi is still in its infancy. Perkins et al., for example, discovered com- pounds of the quinazolinone and phthalazinone structure with modest selectivity for PARPl and PARP2, respec- tively.149 Distinct binding modes necessary for discrimination between Iigands and each homologue were identified, enabling the synthesis of inhibitors with some selectivity for PARPl and PARP2, respectively.63.1S0.151 Moreover, the PARP inhibi- tor BYK204l65 demonstrated lOO-fold selectivity for PARPl over PARP2 in vitro, with IC50 values for PARPl in the nanomolar range.152 Such results demonstrate the feasibility of designing P ARP-homologue-selective Iigands. Furthermore, the recent structural analyses of PARP2 and PARP3 in com- plex with different P ARP inhibitors should be instrumental for the design of homologue-specific inhibitors. 15.1

In summary, since the discovery of poly(ADP-ribosyl)a- tion about half a century ago, the field made significant pro- gress in the understanding of the biochemistry and the cellu- lar function of this post-translational modification. Although there are still a lot of open questions to address, especially regarding the exact cellular functions of the individual P ARPs, it is quite obvious that several P ARPs exhibit impor- tant roles in the process of carcinogenesis. This knowledge can be and is being exploited for establishing new rational treatment modalities for cancer.

1. Hanahan D. Weinberg RA. The hallmarks poly{ADP-ribose)-associated protein ATP-dependent chromatin remodeler.

of cancer. Cell 2000; 100:57-70. complexes. Nucleic Acids Res 2008;36: Proc Natl Acad Sci USA 2009;106:

2. Hoeijmakers JH. DNA damage, aging, 6959-76. 13770-4.

and cancer. N Engl J Med 2009;361: 6. Malanga M, Althaus FR. The role of 9. Ahel D, Horejsi Z. Wiechens N,

1475-85. poly{ADP-ribose) in the DNA damage Polo SE, Garcia-Wilson E. Ahel I,

3. Madhusudan S, Middleton MR. The signaling network. Biochem Cell Bioi 2005; Flynn H, Skehel M, West SC,

emerging role of DNA repair proteins as 83:354-64. Jackson SP, Owen-Hughes T,

predictive, prognostic and therapeutic 7. Pleschke JM, Kleczkowska HE, Strohm M, Boulton SJ. Poly{ADP-ribose)-dependent targets in cancer. Cancer Treat Rev 2005; Althaus FR. Poly{ADP-ribose) binds to regulation of DNA repair by the

31:603-17. specific domains in DNA damage chromatin remodeling enzyme ALC1.

4. Burkle A. Poly{ADP-ribose). The most checkpoint proteins. J Bioi Chem 2000; Science 2009;325:1240-3.

elaborate metabolite of NAD+. FEBS J 275:40974-80. 10. Timinszky G. Till S, Hassa PO, Hothorn 2005;272:4576-89. 8. Gottschalk A], Timinszky G, Kong SE, Jin M, Kustatscher G, Nijmeijer B, 5. Gagne JP, Isabelle M, Lo KS, Bourassa S, J. Cai Y, Swanson SK, Washburn MP, Colombelli J. Altmeyer M, Stelzer EH,

Hendzel MJ, Dawson VL, Dawson TM, Florens L, Ladurner AG, Conaway JW, Scheffzek K, Hottiger MO, Ladurner AG.

Poirier GG. Proteome-wide identification Conaway RC. Poly{ADP-ribosyl)ation A macrodomain-containing histone of poly{ADP-ribose) binding proteins and directs recruitment and activation of an rearranges chromatin upon sensing

(11)

PARPl activation. Nat StYllct Mol Bioi SP. Functions of poly(ADP-ribose) usefulness. Cancer Lett 1981;14:

2009;16:923-9. polymerase in controlling telomere length 227-36.

Il. Ahel I, Ahel D, Matsusaka T, Clark Aj, and chromosomal stability. Nat Genet 37. Staibano S, Pepe S, Lo Muzio L, Somma Pines j, Boulton Sj, West Se. Poly(ADP- 1999;23:76-80. P, Mascolo M, Argenziano G, Scalvenzi ribose)-binding zinc finger motifs in DNA 25. Chang P, Jacobson MK, Mitchison Tj. M, Salvatore G, Fabbrodni G, Molea G, repair/checkpoint proteins. Nature 2008; Poly(ADP-ribose) is required for spindle Bianco AR, Carlomagno C, et al.

451:81-5. assembly and structure. Nature 2004;432: Poly(adenosine diphosphate-ribose)

12. Eustermann S, Brockmann C, Mehrotra 645-9. polymerase 1 expression in malignant

PV, Yang jC, Loakes D, West Se, Ahel I, 26. Kanai M, Tong WM, Sugihara E, Wang melanomas from photoexposed areas of Neuhaus D. Solution structures of the two ZQ, Fukasawa K, Miwa M. Involvement the head and neck region. Hum Pathol PBZ domains from human APLF and of poly(ADP-ribose) polymerase 1 and 2005;36:724-31.

their interaction with poly(ADP-ribose). poly(ADP-ribosyl)ation in regulation of 38. Aguilar-Quesada R, Munoz-Gamez jA, Nat Struct Mol Bioi 2010;17:241-3. centrosome function. Mol Cell Bioi 2003; Martin-Oliva D, Peralta-Leal A, Quiles-

13. Min W, Wang ZQ. Poly (ADP-ribose) 23:2451-62. Perez R, Rodriguez-Vargas jM, Ruiz de

glycohydrolase (PARG) and its 27. Kanai M, Hanashiro K, Kim SH, Hanai S, Almodovar M, Conde C, Ruiz-Extremera therapeutic potential. Front Biosci 2009; Boulares AH, Miwa M, Fukasawa K. A, Oliver Fj. Modulation of transcription

14:1619-26. Inhibition of Crml-p53 interaction and by PARP-l: consequences in

14. Ame jC, Spenlehauer C, de Murcia G. nuclear export of p53 by poly(ADP- carcinogenesis and inflammation. Curr The PARP superfamily. BioEssays 2004;26: ribosyl)ation. Nat Cell Bioi 2007;9: Med Chem 2007;14:1179-87.

882-93. 1175-83. 39. Mangerich A, Scherthan H, Diefenbach j,

15. Hottiger MO, Hassa PO, Luscher B, 28. Wesierska-Gadek j, Ranftler C, Schmid G. Kloz U, van der Hoeven F, Beneke S, Schuler H, Koch-Nolte F. Toward a Physiological ageing: role of p53 and Biirkle A. A caveat in mouse genetic unified nomenclature for mammalian PARP-l tumor suppressors in the engineering: ectopic gene targeting in ES ADP-ribosyltransferases. Trends Biochem regulation of terminal senescence. J cells by bidirectional extension of the Sci 2010;35:208-19. Physiol Pharmacol 2005;56 (Suppl 2): homology arms of a gene replacement

16. Kraus WL. Transcriptional control by 77-88. vector carrying human PARP-l.

PARP-l: chromatin modulation, 29. Meyer-Ficca ML, Meyer RG, Jacobson EL, Transgenic Res 2009;18:261-79.

enhancer-binding, coregulation, and Jacobson MK. Poly(ADP-ribose) 40. Mangerich A, Herbach N, Hanf B, insulation. Curr Opin Cell Bioi 2008;20: polymerases: managing genome Fischbach A, Popp 0, Moreno-Villanueva 294-302. stability. Int J Biochem Cell Bioi 2005;37: M, Bruns OT, Biirkle A. Inflammatory

17. Caiafa P, Guastafierro T, Zampieri M. 920-6. and age-related pathologies in mice

Epigenetics: poly(ADP-ribosyl)ation of 30. Shall S, de Murcia G. Poly(ADP-ribose) with ectopic expression of human PARP-l regulates genomic methylation polymerase-I: what have we learned from PARP-1. Mech Ageing Dev 2010;131:

patterns. Faseb J 2008;23:672-8. the deficient mouse model? Mutat Res 389-404.

18. David KK, Andrabi SA, Dawson TM, 2000;460:1-15. 41. Yelamos j, Schreiber V, Dantzer F.

Dawson VL. Parthanatos, a 31. Tong WM, Cortes U, Wang ZQ. Toward specific functions of poly(ADP- messenger of death. Front Biosci 2009; 14: Poly(ADP-ribose) polymerase: a guardian ribose) polymerase-2. Trends Mol Med

1116-28. angel protecting the genome and 2008;14:169-78.

19. Cohen-Armon M. PARP-l activation in suppressing tumorigenesis. Biochim 42. Menissier de Murcia j, Ricoul M, Tartier the ERK signaling pathway. Trends Biophys Acta 2001;1552:27-37. L, Niedergang C, Huber A, Dantzer F, Pharmacol Sci 2007;28:556-60. 32. Piskunova TS, Yurova MN, Ovsyannikov Schreiber V, Ame jC, Dierich A, LeMeur 20. Haince jF, Kozlov S, Dawson VL, Dawson AI, Semenchenko AV, Zabezhinski MA, M, Sabatier L, Chambon P, et al.

TM, Hendzel Mj, Lavin MF, Poirier GG. Popovich IG, Wang ZQ, Anisimov VN. Functional interaction between PARP-l ATM signaling network is modulated by Deficiency in poly(ADP-ribose) and PARP-2 in chromosome stability and a novel PAR-dependent pathway in the polymerase-l (PARP-I) accelerates aging embryonic development in mouse. Embo J early response to DNA damaging agents. and spontaneous carcinogenesis in mice. 2003;22:2255-63.

J Bioi Chem 2007;282:16441-53. Curr Gerontol Geriatrics Res 2008;2008: 43. Nicolas L, Martinez C, Baro C, Rodriguez

21. Ghodgaonkar MM, Zacal N, Kassam S, 754190. M, Baroja-Maw A, Sole F, Flores jM,

Rainbow Aj, Shah GM. Depletion of 33. Miwa M, Masutani M. PolyADP- Ampurdanes C, Dantzer F, Martin- poly(ADP-ribose) polymerase-l reduces ribosylation and cancer. Cancer Sci 2007; Caballero j, Aparicio P, Yelamos j. Loss

host cell reactivation of a UV -damaged 98:1528-35. of poly(ADP-ribose) polymerase-2 leads

adenovirus-encoded reporter gene in 34. Tomoda T, Kurashige T, Moriki T, to rapid development of spontaneous T- human dermal fibroblasts. DNA Repair Yamamoto H, Fujimoto S, Taniguchi T. cell lymphomas in p53-deficient mice.

(Amst) 2008;7:617-32. Enhanced expression of poly(ADP-ribose) Oncogene 2010;29:2877-83.

22. Flohr e, Biirkle A, Radicella JP, Epe B. synthetase gene in malignant lymphoma. 44. Rouleau M, McDonald D, Gagne P, Poly(ADP-ribosyl)ation accelerates DNA Am J HematoI1991;37:223-7. Ouellet ME, Droit A, Hunter jM, repair in a pathway dependent on 35. Shiobara M, Miyazaki M, Ito H, Togawa Dutertre S, Prigent C, Hendzel Mj, Cockayne syndrome B protein. Nucleic A, Nakajima N, Nomura F, Morinaga N, Poirier GG. PARP-3 associates with Acids Res 2003;31 :5332-7. Noda M. Enhanced polyadenosine polycomb group bodies and with 23. Beneke S, Cohausz 0, Malanga M, diphosphate-ribosylation in cirrhotic liver components of the DNA damage repair

Boukamp P, Althaus F, Biirkle A. Rapid and carcinoma tissues in patients with machinery. J Cell Biochem 2007; 100:

regulation of telomere length is mediated hepatocellular carcinoma. J Gastroenterol 385-401.

by poly(ADP-ribose) polymerase-I. HepatoI2001;16:338-44. 45. Loseva 0, jemth AS, Bryant HE, Schuler Nucleic Acids Res 2008;36:6309-17. 36. Fukushima M, Kuzuya K, Ota K, Ikai K. H, Lehtio L, Karlberg T, Helleday T.

24. d'Adda di Fagagna F, Hande MP, Tong Poly(ADP-ribose) synthesis in human Poly(ADP-ribose) polymerase-3 (PARP-3) WM, Lansdorp PM, Wang ZQ, jackson cervical cancer cell-diagnostic cytological is a mono-ADP ribosylase that activates

Referenzen

ÄHNLICHE DOKUMENTE

Efalizumab inhibits the binding of αLβ2 to ICAM-1, -2, and -3 (Boehncke 2007a), and it is also able to inhibit the mixed lymphocyte response, T cell

Antimalarial activity of inhibitors of ADP-ribosyl transferase against Plasmodium yoelii infection in mice.. Preliminary data Short

To address this question, we measured cellular poly(- ADP-ribosyl)ation in PBMC from elderly donors as a function of plasma zinc concentrations before and after a 7-week course of

In addition, the presence of PARP1 at the silenced p16/INK4 promoter in the absence of CTCF (Witcher and Emerson, 2009) argues in favor of the hypothesis that PARP1 independently

The next day, cells were treated with or without 1 mM H 2 O 2 for 7 min at 37uC and fixed either with 100% methanol for 7 min at 4uC, with formaldehyde at varying concentrations

Effect of rapamycin on the automodification activity of recombinant purified PARP-1 in vitro and in the presence of varying activator oligonucleotide concentrations.. (A) PARP-1

Poly(ADP-ribosyl)ation is involved in a variety of cellular functions such as the regulation of DNA repair, genomic stability, longevity, transcription, cell

In summary, ex vivo supplementation of human PBMC with NA increases cellular NAD+ levels, boosts the cellular poly(ADP-ribosyl)ation response to genotoxic