Translational Chemotherapy for triple negative Breast Cancer - A Review on significance of poly (ADP-ribose) polymerase 1 (PARP 1) inhibitors

 

Elizabeth Eldhose1, B. Gowramma1*, Manal Mohammed2, R. Kalirajan1, L. Kaviarasan1

1Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Udhagamandalam, Tamilnadu,

2Department of Pharmaceutical Chemistry, KTN College of Pharmacy, Kerala.

*Corresponding Author E-mail: gowrammab@rediffmail.com, gowrammab@jssuni.edu.in

 

ABSTRACT:

Breast Cancer, the most common cancer observed in women around the world[1], accounts for 12% of all new cancer cases and nearly 25% of all cancers in women[2]. Breast Cancer, a heterogenous disease, is evident over a broad differentiation in phenotypes and morphological profiles, with an after effect of various clinical behaviours[3]. From an estimated 1 million breast cancer cases diagnosed worldwide, 170,000 are of triple negative phenotype (15-20%)[4]. Triple Negative Breast Cancer (TNBC) is a substantially histopathological category based, where there is deficiency of expression of hormone receptors (ER and PR) as well as no transmutation of human epidermal growth factor receptor type 2 (HER2)[3]. They are characterized by poor prognosis and aggressiveness construed by low five-year survival and high recurrence rates after adjuvant therapy. TNBC share arresting correlation with basal-like breast cancers. It is observed with high frequency of BRCA1 mutations and a heightened Ki-67 expression. Taxanes and/or platinum compounds and PARP 1 inhibitors are a good choice of treatment for TNBC[4]. The intent of this review is to highlight the relevance of PARP 1 inhibitors on TNBC and display an in depth discussion regarding these futuristic inhibitors.

 

KEYWORDS: Breast Cancer, Triple Negative Breast Cancer, Human Epidermal Growth Factor Receptor, Taxanes, Poly (ADP-ribose) polymerase 1 Inhibitor.

 

 


INTRODUCTION:

TNBC is now one of the most active areas of research in oncology. A patient suffering from this subset of breast cancer undergoes an extremely testing and thwarting situation. TNBC, a heterogenous breast cancer subdivision, shows only a limited overlying with the so-called basal breast cancer. Hence it can be said that “triple negative” and “basal-like” are not fully interchangeable. It has to be kept in mind that not all TNBC are of basal-like subtype and not all basal-like breast cancer is triple negative. By definition, triple negative implies to the immune histo-chemical classification of breast tumours lacking ER, PR and HER2 expression although the basal-like subdivision is explained via gene expression microarray analysis[5,6].

 

 

 

Currently, there are six sub-types of TNBC which includes basal-like 1(BL-1), basal-like 2(BL-2), immunomodulatory (IM), mesenchymal (M), mesenchymal stem-like (MSL) and luminal androgen receptor (LAR)-positive[7].

 

Gene Expression profiling has proved that relatively 80% of all TNBC tumours are basal-like[8]. Basal-like TNBC can be indicated by noticeable risk factors: aggressive and early The Gene Expression profiling has proved that relatively 80% of all TNBC tumours are basal-like[6]. Basal-like TNBC can be indicated by noticeable risk factors: aggressive and early patterns of metastases, exclusive molecular characteristics, association with BRCA1 mutations, corresponding lack of targeted therapeutics and poor prognosis and patient survival rate[9-16]. The BL-1 sub-type is outlined by an enriched cell cycle and DNA damage response gene expression[7]. Enhancement in proliferation genes and expanded Ki-67 expression in basal-like TNBC can be the reason why this type responded well to antimitotic agents such as taxanes and DNA damaging agents like Cisplatin[17,18].

 

Clinical attributes of TN tumors includes markedly elevated mitotic count, axillary node involvement, tumour necrosis, pushing margin of invasion and stromal lymphocytic response and high nuclear-cystoplasmic ratio[19,20]. These tumours are histologically ductal invasive carcinomas characterized by poor differentiation, high proliferative capacity and overall large tumour size, but metaplastic and medullary histologies are also observed[21]. It was observed that these tumours frequently disseminate to the lungs and the brain tissue which is contradictory to what is observed in other subtypes of breast cancer where metastasis occurs in bone and soft tissue[22-24]. Furthermore, the entrenched correlation between tumour size and positive lymph nodes which has been detected in other breast cancer subdivisions is not pertinent to TN tumours[25]. TNBC is more rampant in women of African-American and Hispanic origin and in younger women (age less than 40)[9,23]. The first five year survival rate in TNBC is predicted at 70% which is lower than the 80% observed in other subdivisions.

 

POLY (ADP-RIBOSE) POLYMERASES 1 (PARP-1):

Poly (ADP-Ribose) Polymerases constitute a clan of enzymes which are liable for the catalyzed transfer of ADP-ribose moieties onto specific acceptor proteins and transcription factors, utilizing nicotine adenine nucleotide (NAD+) as the substrate[26,27]. They consist of linear and/or branched repeats of ADB-ribose, which can be long up to 200 units.

 

PARP1 which is the most abundant, ubiquitously expressed nuclear protein is involved in the mediation of DNA damage response (at this stage, its enzymatic activity was observed to increase about 500-fold) and is the best defined isoform of the PARP family[28]. It is accountable for the 85-90% of the total poly (ADP-ribosylation) activity and is localized within the cell nucleus[29]. It binds with the DNA via two zinc finger motifs and relocates chains of ADP-ribose moieties from NAD+ to the chromatin-associated acceptor proteins. The covalent modifications catalyzed by the PARP1 is responsible for the regulation of the activity of proteins involved in the regeneration of DNA damages and the preservation of genome stability, gene transcription, proliferation, and differentiation of cells, and other processes[30,31]. PARP1 plays an active role in in several biological mechanisms counting hypoxic response, transcriptional regulation, maintenance of chromosome stability, DNA repair and apoptosis[32,33]. It principally operates as a DNA repair factor, chiefly in base excision repair. Other therapeutic areas save for cancer, where PARP1 is involved are stroke, inflammation, cardiac ischemia and diabetes[34-37]. Accelerated binding of the PARP1 with the DNA strand breaks is essential for the resealing of the DNA single strand breaks during base excision repair and for the overhaul of DNA double strand breaks and topoisomerase I cleavage complexes. The gravity of PARP1 in cancer was further reinforced by two recent seminal preclinical studies[38,39] where it was unveiled that the cell lines deficient in BRCA1 and BRCA2 (faulty in homologous recombination repair) were 1000 times more susceptible to PARP inhibition compared to the wild-type or heterozygous mutant cells. This discovery accentuated that PARP1 is the synthetic lethal ally of BRCA1 and BRCA2 and inhibition of PARP1 can prompt vastly scrupulous killing of BRCA1/2-deficient tumour cells[36]. PARP1 has an indispensable role in the repair of single strand breaks by the base excision repair pathway.

 

The PARP1 enzyme (Figure 1) is a 113kDa protein accompanying three primary structural domains, a DNA binding domain with two zinc fingers, a 55kDa catalytic domain which employs NAD+ as the substrate to shape polymers of ADP-ribose on histones and the additional acceptor nuclear protein encompass the automodification domain of PARP1 itself[40-42]. NAD+ binding site of PARP-1 contains two distinctive binding zones, specifically, a nicotinamide–ribose binding site (NI site) and an adenine–ribose binding site (AD site)[43].

 

Figure 1: The PARP1 enzyme accompanying three primary structural domains

 

DNA damage generated by peroxidation[44], irradiation[45], and DNA damaging chemicals like chemotherapeutic agents[46] prompt the catalytic activity of PARP-1. Subsequently to DNA damage, the 42kDa DNA binding domain with two zinc fingers binds with the damaged DNA and triggers polymerization of ADP-ribose culminating in the unfolding of DNA from histones and disclosing the damaged DNA for repair[41]. The 2’-OH of the substrate NAD+ binds to an intermediate oxonium ion which is generated by the detachment of nicotinamide from the ribose ring. A hydrogen bonding network is formed between critical amino acid residues which includes Ser904 and Gly863 with the nicotinamide moiety[46]. Further, Tyr904 forms a planar surface which develops a pi-pi interaction with the nicotinamide moiety and maintains the oxonium ion. Simultaneously, Glu988 expedites the proton transfer from the 2’-OH group before the glycosidic bond is established[47]. The synthesis of branched polymers from the 2’-OH of the nicotinamide ribose is also mobilized by PARP1.

 

MECHANISMS OF PARP1 INHIBITORS FOR ANTICANCER ACTIVITY:

PARP1 inhibition (Figure 2) when considered as a target for oncology, acts as a chemopotentiator considering the fact that most of the anticancer therapeutics focus on DNA damage as a tool to eradicate the expeditiously splitting cancer cells. DNA repair for abundant cancerous cell types is carried out by the PARP1 mediated repair pathway[48]. Therefore, inhibition of PARP1 along with DNA damaging chemotherapeutics or radiation is a very competent tool to jeopardize the cancer cell DNA repair mechanism, ensuing genomic dysfunction and cell death.

 

Figure 2: Mechanisms of PARP1 Inhibitors for Anticancer Activity

 

Breast Cancer linked genes BRCA and BRCA2 have been distinguished hitherto as tumor suppressor genes that play a indispensable part in the repair of double strand breaks (DSB) in DNA via a process called homologous recombination[49]. Albeit PARP1 inhibition will contribute to an escalation in single strand breaks (SSB), the prevalence of these SSBs will ultimately lead to DSBs through replication fork collapse[50]. The hike of DSBs in the existence of HR lacking cell types causes chromosomal aberrations and vulnerability of the genome resulting in cell death. This incident is referred to as synthetic lethality[51], specifically since the loss of one gene function will result in cell susceptibility (i.e., the loss of PARP1 or BRCA1/2) but the loss of both is fatal (i.e., BRCA1/2 deficient cells and PARP1 inhibitor) [52].

 

Another mechanism of PARP1 inhibition exerting anticancer property, involves the interactions between DNA, PARP1 and its inhibitors which results in inhibition of PARP1 catalytic activity and ambush of the DNA-PARP1 complexes[53]. Noncovalent DNA-PARP complexes occur normally in intact cells. When PARP1 inhibitors bind to the NAD+ binding pocket of the PARP1, an allosteric conformational change is promoted and reversible association of PARP1/2 with DNA is preserved. This mechanism is termed as the trapping of DNA-PARP1/2 complexes[52,54-56]. Methodical observations[56] reveals that the DNA-PARP trapping ability of PARP1 inhibitors has a much greater correlation with their cytotoxicity than their capability to suppress PARP1 catalytic activity. Levels of DSBs are more significantly correlated with cell death than are levels of trapping. Cell killing effect of this entrapment, are generally through the transformation of unrepaired SSBs into lethal DSBs.

 

Categories Of Parp1 Inhibitors:

PARP1 inhibitors target the NAD+ binding site by occupying the nicotinamide pocket. The first discovered inhibitors 3-amino/nitrobenzamides (Figure 3) which is structurally analogous to the PARylation by-product nicotinamide[57]. These compounds showed low-to-moderate activity against PARP1 and non-optimal PK properties. Second generation of PARP1 inhibitors were more effective and target specific and were developed based on quinazoline analogues (specifically, 1,5-dihydroisoquinoline)[58]. In fact, development of various newer drug groups was based on these molecules. Majority of the rest PARP1 inhibitors are bicyclic and tricyclic compounds where the pivotal carboxamido moiety is buried inside a ring system to form a lactam. Meanwhile an ancillary appendage with a shorter or longer linking chain is usually adhered to the polycyclic core as a solvent accessory region, which is generally required to connect the NI binding and AD binding units[59-61]. This chain is also paramount for the potency and the physicochemical properties of the inhibitors.

 

Figure 3: The first discovered inhibitors 3-amino/nitrobenzamides

 

Nowadays, generally two types of PARP1 inhibitors are observed, consistently endowed with two distinguishing nicotinamide-mimic motifs (1) a rotationally constrained primary amide, in particular clinical candidates like Veliparib (Abbott)[62] and Niraparib (Merck)[63] or (2) an amide embedded in a ring as in Rucaparib (Pfizer/ Clovis)[64], Olaparib (Astra Zeneca)[65] now marketed as Lynparza and Talaxoparib (BioMarin)[66]. The figure 4 shows above mentioned PARB inhibitors. From the PARP family of enzymes, none of the above mentioned inhibitors selectively inhibits PARP1. For example, all the clinical candidates and extensive majority of stated inhibitors also interact with PARP 2 with similar potencies. PARP-2 (aka ARTD-228) which is a 62 kDa nuclear protein and similar to PARP-1, is involved in DNA single-strand break repair. Yet, its input is nominal (5 −10%) to the total DNA damage induced PARP activity[67].

 

 

Figure 4: PARB inhibitors

 

Olaparib, a robust oral PARP inhibitor is fatal to cells accommodating BRCA1 or BRCA2 mutations. During Phase I trial, the pharmcokinetic and pharmacodynamic parameters of the olaparib capsule formulation was established by Fong et al. where it was determined that pharmacokinetic characteristics at twice-daily dosing was rapid absorption and elimination[55]. The peak plasma concentration of Olaparib was attained within 1-3 h after its intake, following a biphasic slump in plasma concentrations with a terminal elimination half-life of 5-7 h. Olaparib is mainly metabolized via dehydrogenation and oxidation, with a number of components which are further metabolized by glucuronide or sulphate conjugation. Cytochrome P450 (CYP) 3A4 is the principal metabolizing enzyme of olaparib, hence coadministration of olaparib with strong or moderate CYP3A4 inhibitors or inducers is generally avoided. Ang et al carried out a mass balance study of olaparib and established that excretion of the drug occurs primarily via faeces (42%) and urine (44%).

 

 

Interactions of PARP1 with its inhibitor:

H-bonding networks formed between the carboxamido component and Ser904 and Gly863 of PARP1, and the π–π stacking between the aryl ring and Tyr907 were the interactions observed in the 3-amino/nitrobenzamides, the first inhibitors[59]. All of the known PARP1 inhibitors showed interactions via molecular modelling studies, with the NI site through three vital hydrogen bonds formed between the lactam or the carboxamide group of the small molecules and residues Ser904 and Gly863 of the catalytic site[68,69]. (Figure 5)

 

 

Figure 5: Interactions of PARP1 with its Inhibitor

 

Furthermore, a pi-pi stacking interaction between the aromatic scaffold of PARP1 inhibitors and Tyr 896 and Tyr 907 of the NI site was customarily detected as  additional fundamental feature. The AD site is quite large when compared with the nearly cramped NI site and can be used to search for an extensive range of novel inhibitors with the enhanced potency and pharmacokinetic properties[70].

 

Cyclic inhibitors which are conformationally constrained establish an anti-disposition of the amide bond. Improved PARP inhibitor’s affinity to the binding site can be observed by locking the carboxamide moiety which is usually free to rotate. This locking process can be done by either introducing heteroatoms or groups on the aromatic ring which are able to provide an intramolecular hydrogen bond with the amide NH, or encompassing the amide group into a two (or more) ring heterocycle[59].

 

Analysing these findings, a pharmacophore query was created by Giannini et al. where presence of three structural features were enforced (a) an aromatic ring, (b) a carboxamide moiety with at least one NH group into the desired anti-conformation, and (c) a side chain extending into the deep cavity placed in the auto-modification domain of PARP1.

 

Recent study on novel scaffold for PARP1 inhibitior:

Fu et al discovered a series of dihydrodibenzo[b,e]-oxepin compounds as PARP1 inhibitors. Lead Optimization resulted in the identification of compound (2-(11-(3-(dimethylamino)propylidene)-6,11-dihydrodibenzo[b,e] oxepin)-2yl)acetohydrazide), which showed a novel chemical scaffold and exclusive binding interactions with PARP1 protein. This particular compound exhibited excellent potency (inhibiting PARP1 enzyme activity with IC50 = 0.079μM) as well as inhibiting PARP-modulated PARylation and cell proliferation in MDA-MB-436 cells (BRCA1 mutation). Further, the compound also inhibited cell migration that is closely associated to cancer metastasis and presented exceptional anti-tumor efficacy in MDA-MB-436 xenograft model without apparent toxicity. Structure based Pharmacophore detection led to four common features on comparison, comprising A1 (hydrogen bond acceptor), D1 (hydrogen bond donor), AR1 (ring aromatic) and H1(hydrophobic). Extensive Structural Activity Relationship (Figure 6) was operated after recognising the most potent compound. The scaffold was classified into three rings. The activity of seven membered rings in Part A was found to be remarkable on comparison with the six membered ring, and when the substituent X is O, the activity was better. In Part B, higher activity was observed when R2 was substituted for different amide groups. Carbon chain requires a certain length in Part C and the best activity was noticed when R1 was substituted with tertiary amine groups.

 

 

Figure 6: Structural Activity Relationship for Novel Scaffold for PARP1 Inhibition

 

Clinical relevance of PARP1 inhibitors:

During clinical evaluation of PARP1 inhibitors, the compounds were tested against breast, ovarian or pancreatic cancer accommodating BRCA defects. Along with this investigation, these inhibitors along with the traditional chemotherapeutic agents were also studied against other solid tumors, including non small cell lung cancer and glioblastoma. The best response of Olaparib was shown against BRCA-mutated breast cancer. Olaparib was used in combination with DNA damaged agents, microtubule inhibitors, angiogenesis inhibitors and kinase inhibitors during the clinical trials. From six Phase I studies, this compound was acknowledged to improve the therapeutic response of patients with various cancers to cisplatin, carboplatin and pegylated liposomal doxorubicin, a topoisomerase II inhibitor, along with increased toxicities[71-73]. Combination of Olaparib and Cediranib, a vascular endothelial growth factor receptor (VEGFR) inhibitor showed progress in the therapeutic efficacy in patients with ovarian cancer regardless of BRCA stature[74,75]. Gefitinib, an epidermal growth factor receptor and the panphosphatidylinositol 3-kinase (PI3K) inhibitor were found to offer supplementary clinical assistance with use of Olaparib[76,77].

 

CONCLUSION:

PARP1 plays important roles in both DNA repair and transcription, and the interplay of these processes in relation to cellular function and diseases states have not been well defined. As PARP1 binding motifs may be readily found in promoter elements of DNA repair genes, the expanding role of PARP1 in DNA repair need not be independent of transcription. The discovery of ADP-ribose binding modules that bind to various forms mono- and poly- ADP-ribose has provided important insights into how ADP-ribosylation regulates different cellular pathways. Among the four distinct PAR-binding modules discovered so far, it is the macrodomain alone that, in addition to possessing binding activity, in some instances also supports a catalytic activity toward ADP-ribose derivates. However, the development of PARP inhibitors as chemopotentiating agents has been limited by an increase in observed toxicities, mainly myelosuppression, necessitating dose reductions of the cytotoxic chemotherapeutic agent and the PARP inhibitor. Hence it presents an opportunity to rationally develop combinations of PARP inhibitors with new classes of DNA repair inhibitors that are on the horizon, and classical cytotoxic agents. In this reviwe, triple negative breast cancer, PARP1 and inhibitors, its role in TNBC, mechanism, categories, interactions, recent study on novel scaffold and clinical significance of PARP1 inhibitors were deliberated and reviewed.

 

REFERENCES:

1.     World Health Organization, Breast Cancer, Global Health Estimates, Geneva - WHO 2013(http://www.who.int/cancer/detection/breastcancer/en/index1.html)

2.     World Cancer Research Fund, Breast Cancer Statistics 2012 (https://www.wcrf.org/int/cancer-facts-figures/data-specific-cancers/breast-cancer-statistics)

3.     Weigelt B, Baehner FL, Reis Filho, JS. The contribution of gene expression profiling to breast cancer classification, prognostication and prediction: a retrospective of the last decade. J. Pathol 2010; 220: 263–280.

4.     Marusyk A, Polyak K. Tumor heterogeneity: causes and consequences. Biochim Biophys Acta 2010; 1805(1): 105–17.

5.     Anders CK, Carey LA. Biology, metastatic patterns and treatment of patients with triple-negative breast cancer. Clin Breast Cancer 2009, 9: 73-81.

6.     Sorlie T, Perou CM, Tibshirani R. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA 2001, 98: 10869 - 10874.

7.     Sorlie T, Tibshirani R, Parker J. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci USA 2003, 100: 8418 - 8423.

8.     Lehmann BD, Bauer JA, Chen X. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin Investig 2011, 121: 2750 – 2767.

9.     Carey L, Perou C, Livasy C, Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA 2006, 295: 2492 – 2502.

10.   Dent R, Trudeau M, Pritchard K. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res 2007, 13: 4429 – 4434.

11.   Livasy CA, Karaca G, Nanda R. Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol 2006, 19: 264 – 271.

12.   Millikan RC, Newman B, Tse CK. Epidemiology of basal-like breast cancer. Breast Cancer Res Treat 2008, 109: 123–39.

13.   Nielsen TO, Hsu F, Jensen K. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res 2004, 10: 5367–74.

14.   Perou CM, Sorlie T, Eisen M.B. Molecular portraits of human breast tumours. Nature 2000, 406: 747–52.

15.   Smid M, Wang Y, Zhang Y. Subtypes of breast cancer show preferential site of relapse. Cancer Res 2008, 68: 3108 – 3114.

16.   Sorlie T, Tibshirani R, Parker J. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A.2003, 100: 8418 – 8423.

17.   Chakravarthy, AB, Kelley MC, McLaren B, Truica CI, Billheimer D, Mayer IA. Neoadjuvant concurrent paclitaxel and radiation in stage II/III breast cancer. Clin Cancer Res 2006, 12(5): 1570 – 1576.

18.   Bauer JA, Chakravarthy AB, Rosenbluth JM, Mi D, Seeley EH, De Matos GIN. Identification of markers of taxane sensitivity using proteomic and genomic analyses of breast tumors from patients receiving neoadjuvant paclitaxel and radiation. Clin Cancer Res 2010, 16(2): 681– 690.

19.   Bertucci F, Finetti P, Cervera N. How basal are triple-negative breast cancers. Int J Cancer 2008, 123: 236–240.

20.   Rakha EA, Elsheikh SE, Aleskandarany, MA. Triple-negative breast cancer: distinguishing between basal and nonbasal subtypes. Clin Cancer Res 2009, 15: 2302–2310.

21.   Livasy, CA, Karaca G, Nanda, R. Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol 2006; 19(2): 264 – 271.

22.   Fulford LG, Easton DF, Reis Filho JS. Specific morphological features predictive for the basal phenotype in grade 3 invasive ductal carcinoma of breast. Histopathology 2006; 49(1): 22–34.

23.   Huo D, Ikpatt F, Khramtsov A, Dangou JM. Population differences in breast cancer: survey in indigenous African women reveals over-representation of triple-negative breast cancer. J Clin Oncol 2010; 28: 3271 – 3277.

24.   Kennecke H, Yerushalmi R, Woods R, Cheang MC, Voduc D, Speers CH, Nielsen TO, Gelmon K. Metastatic behavior of breast cancer subtypes. J. Clin Oncol 2010; 28, 3271 – 3277.

25.   Sihto H, Lundin J, Lundin M, Lehtimaki T, Ristimaki A, Holli K, Sailas L, Kataja V, Turpeenniemi Hujanen T, Isola J, Heikkila P, Joensuu H. Breast cancer biological subtypes and protein expression predict for the preferential distant metastasis sites: a nationwide cohort study. Breast Cancer Res 2011; 13: 87.

26.   Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG. Nat. Rev.Cancer 2010; 10: 293.

27.   Chambon P, Weil JD, Mandel P. Biochem. Biophys. Res. Commun 1963; 11.

28.   Hottiger MO, Hassa PO, Lüscher B, Schuuler H, Koch Nolte F. Toward a unified nomenclature for mammalian ADP-ribosyltransferases. Trends Biochem. Sci 2010; 35: 208−219.

29.   De Vos M, Schreiber V, Dantzer F. Biochem. Pharmacol. 2012; 84: 137.

30.   Hassa PO, Haenni SS, Elser M, Hottiger MO.  Microbiol. Mol. Biol. Rev 2006; 70: 789.

31.   Hassa, P. O.; Hottiger, M. O.; Front. Biosci., 2008, 13, 3046.

32.   Schreiber V, Dantzer F, Ame JC, Murcia G. Poly (ADP-ribose): novel functions for an old molecule. Nat. Rev. Mol. Cell Biol 2006; 7: 517–528.

33.   Weaver AN, Yang ES. Beyond DNA repair: additional functions of PARP1 in cancer. Front. Onco 2013; 3; 290.

34.   Bai P, Canto C. The role of PARP-1 and PARP-2 enzymes in metabolic regulation and disease. Cell Metab 2012; 16: 290–295.

35.   Gibson BA, Kraus WL. New insights into the molecular and cellular functions of poly (ADP-ribose) and PARPs. Nat. Rev. Mol. Cell Biol 2012; 13: 411–424.

36.   Horvath EM, Szabo C. Poly (ADP-ribose) polymerase as a drug target for car-diovascular disease and cancer: an update. Drug News Perspect 2007; 20: 171–181

37.   De la Lastra, C.A.; Villegas, I.; Sanchez Fidalgo, S. Poly (ADP-ribose) polymeraseinhibitors: new pharmacological functions and potential clinical implications. Curr. Pharm. Des 2007; 13: 933 – 962.

38.   Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, Kyle S, Meuth M, Curtin NJ, Helleday T. Specific killing of BRCA2-deficient tumours with inhibitors of poly (ADP-ribose) polymerase. Nature 2005; 434: 913-917.

39.   Farmer H, Mc Cabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, Santarosa M, Dillon KJ, Hickson I, Knights C, Martin NM, Jackson SP, Smith GC, Ashworth A. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005; 434: 917-921.

40.   Sandhu SK, Yap TA, De Bono JS. Poly (ADP-ribose) polymerase inhibitors in cancer treatment: a clinical perspective. Eur. J. Cancer 2010; 46: 9-20.

41.   Langelier MF, Servent KM. Rogers EE, Pascal JM. A third zinc-binding domain of human poly(ADP-ribose) polymerase-1 coordinates DNA-dependent enzyme activation. J. Biol. Chem 2008; 283: 4105 - 4114.

42.   Langelier MF, Ruhl DD, Planck JL, Kraus WL, Pascal JM. The Zn3 domain of human poly (ADP-ribose) polymerase-1 (PARP-1) functions in both DNA-dependent poly (ADP-ribose) synthesis activity and chromatin compaction. J. Biol. Chem 2010; 285: 18877-18887

43.   Ishida J, Yamamoto H, Kido Y, Kamijo K, Murano K, Miyake H, Ohkubo M, Kinoshita T, Warizaya M, Iwashita A, Mihara K, Matsuoka N, Hattori K. Bioorg. Med. Chem 2006; 14: 1378.

44.   Szabo C, Zingarelli B, Connor M, Salzman AL. DNA strand breakage, activation of poly (ADP-ribose) synthetase, and cellular energy depletion are involved in the cytotoxicity of macrophages and smooth muscle cells exposed to peroxynitrite. Proc. Natl. Acad. Sci. U.S.A 1996; 93: 1753–1758.

45.   De Murcia JM, Niedergang C, Trucco C, Ricoul M, Dutrillaux B, Mark M, Oliver FJ, Masson M, Dierich A, Le Meur M, Walztinger C, Chambon P, De Murcia G. Requirement of poly (ADP-ribose) polymerase in recovery from DNA damage in mice and in cells. Proc. Natl. Acad. Sci. U.S.A. 1997; 94: 7303–7307.

46.   Bowman KJ, White A, Golding BT, Griffin RJ, Curtin NJ. Potentiation of anti-cancer agent cytotoxicity by the potent poly (ADP-ribose) polymerase inhibitors NU1025 and NU1064. Br. J. Cancer 1998; 78: 1269–1277.

47.   Ruf A, De Murcia G, Schulz GE. Inhibitor and NAD+ binding to poly (ADP-ribose) polymerase as derived from crystal structures and homology modeling. Biochemistry 1998; 37: 3893– 3900.

48.   Ljungman M. Targeting the DNA damage response in cancer. Chem. Rev 2009; 109: 2929–50.

49.   Lord CJ, Ashworth A. Targeted therapy for cancer using PARP inhibitors. Curr. Opin. Pharmacol 2008; 8: 363–369.

50.   Saffhill R, Ockey CH. Strand breaks arising from the repair of the 5-bromodeoxyuridine-substituted template and methyl methanesulphonate-induced lesions can explain the formation of sister chromatid exchanges. Chromosoma 1985; 92: 218–224.

51.   Kaelin WG, The concept of synthetic lethality in the context of anticancer therapy. Nat. Rev. Cancer, 2005; 5: 689–698.

52.   Shen Y, Aoyagi Scharber M, Wang B. Trapping poly (ADPRibose) polymerase. J. Pharmacol. Exp. Ther 2015; 353: 446−457.

53.   Scott CL, Swisher EM, Kaufmann SH. Poly (ADP-ribose) polymerase inhibitors: recent advances and future development. J. Clin. Oncol 2015; 33: 1397−1406.

54.   Murai J, Huang SY, Das BB, Renaud A, Zhang Y, Doroshow JH, Ji J, Takeda S, Pommier Y. Trapping of PARP1 and PARP-2 by clinical PARP inhibitors. Cancer Res, 2012; 72: 5588− 5599.

55.   Murai J, Huang SY, Renaud A, Zhang Y, Ji J, Takeda S, Morris J, Teicher B, Doroshow JH, Pommier Y. Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Mol. Cancer Ther 2014; 13: 433−443.

56.   Murai J, Zhang Y, Morris J, Ji J, Takeda S, Doroshow JH, Pommier Y. Rationale for poly (ADP-ribose) polymerase (PARP) inhibitors in combination therapy with camptothecins or Temozolomide based on PARP trapping versus catalytic inhibition. J. Pharmacol. Exp. Ther 2014; 349: 408−416.

57.   Patel AG, De Lorenzo SB, Flatten KS, Poirier GG, Kaufmann SH. Clin. Cancer Res 2012; 18: 1655-1662.

58.   Banasik M, Komura H, Shimoyama M, Ueda K. J. Biol. Chem 1992; 267(3): 1569–1575.

59.   Ferraris DV. Evolution of poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors. From concept to clinic. J. Med. Chem 2010; 53: 4561-4584.

60.   Penning TD. Small-molecule PARP modulators--current status and future therapeutic potential. Curr. Opin. Drug Discov. Devel 2010; 13: 577-586.

61.   Davar D, Beumer JH, Hamieh L, Tawbi H. Role of PARP inhibitors in cancer biology and therapy. Curr. Med. Chem 2012; 19: 3907-3921.

62.   Penning TD, Zhu GD, Gandhi VB, Gong J, Liu X, Shi Y, Klinghofer V, Johnson EF, Donawho CK, Frost DJ, Bontcheva Diaz V, Bouska JJ, Osterling DJ, Olson AM, Marsh KC, Luo Y, Giranda VL. Discovery of the poly-(ADP-ribose) polymerase (PARP) inhibitor 2-[(R)-2-methylpyrrolidin-2-yl]-1Hbenzimidazole- 4-carboxamide (ABT-888) for the treatment of cancer. J. Med. Chem 2009; 52: 514−523.

63.   Jones P, Altamura S, Boueres J, Ferrigno F, Fonsi M, Giomini C, Lamartina S, Monteagudo E, Ontoria JM, Orsale MV, Palumbi MC, Pesci S, Roscilli G, Scarpelli R, Schultz Fademrecht C, Toniatti C, Rowley M. Discovery of 2-{4-[(3S)- piperidin-3-yl]phenyl} -2H- indazole -7- carboxamide (MK-4827): a novel oral poly (ADP-ribose) polymerase (PARP) inhibitor efficacious in BRCA-1 and −2 mutant tumors. J. Med. Chem 2009; 52: 7170− 7185.

64.   Jones, P.; Wilcoxen, K.; Rowley, M.; Toniatti, C. Niraparib: a poly (ADP-ribose) polymerase (PARP) inhibitor for the treatment of tumors with defective homologous recombination. J. Med. Chem., 2015, 58, 3302−3314.

65.   Thomas HD, Calabrese CR, Batey MA, Canan S, Hostomsky Z, Kyle S, Maegley KA, Newell DR, Skalitzky D, Wang LZ. Webber SE, Curtin NJ. Preclinical selection of a novel poly (ADP-ribose) polymerase inhibitor for clinical trial. Mol. Cancer Ther 2007; 6: 945−956.

66.   Menear KA, Adcock C, Boulter R, Cockcroft XL, Copsey L, Cranston A, Dillon KJ, Drzewiecki J, Garman S, Gomez S, Javaid H, Kerrigan F, Knights C, Lau A, Loh VM. Matthews IT, Moore S, Connor MJ, Smith GC, Martin NM. 4-[3-(4-Cyclopropane carbonylpiperazine-1-carbonyl)-4-fluorobenzyl]-2H-phthalazin-1-one: a novel bioavailable inhibitor of poly (ADP-ribose) polymerase-1. J. Med. Chem 2008; 51: 6581−6591.

67.   Shen Y, Rehman FL, Feng Y, Boshuizen J, Bajrami L, Elliott R, Wang B, Lord CJ, Post LE, Ashworth A. BMN673, a novel and highly potent PARP1/2 inhibitor for the treatment of human cancers with DNA repair deficiency. Clin. Cancer Res 2013; 19: 5003−5015.

68.   Shen Y, Aoyagi Scharber M, Wang B. Trapping Poly (ADP-Ribose) Polymerase. J. Pharmacol. Exp. Ther 2015; 353: 446−457.

69.   Yelamos J, Schreiber V, Dantzer F. Toward specific functions of poly (ADP-ribose) polymerase-2. Trends Mol. Med 2008; 14: 169−178.

70.   Ruf, A.; Menissier de Murcia, J.; de Murcia, G.; Schulz, G. E. Proc. Natl. Acad. Sci. U.S.A., 1996, 93, 7481.

71.   Ruf A, De Murcia G, Schulz GE. Biochemistry.1998; 37: 3893.

72.   Lehtio L, Jemth AS, Collins R, Loseva O, Johansson A, Markova N, Hammarstrom M, Flores A, Holmberg Schiavone L, Weigelt J, Helleday T, Schüler H, Karlberg TJ. Med. Chem, 2009; 52: 3108.

73.   Lee JM, Hays JL, Annunziata CM, Noonan AM, Minasian L, Zujewski JA, Yu M, Gordon N, Ji J, Sissung TM, Figg WD, Azad N, Wood BJ, Doroshow J, Kohn EC. Phase I/Ib study of olaparib and carboplatin in BRCA1 or BRCA2 mutation associated breast or ovarian cancer with biomarker analyses. J. Natl. Cancer Inst; 2014: 89, 106.

74.   Balmanna J, Tung NM, Isakoff SJ, Granna B, Ryan PD, Saura C, Lowe ES, Frewer P, Winer E, Baselga J, Garber JE. Phase I trial of olaparib in combination with cisplatin for the treatment of patients with advanced breast, ovarian and other solid tumors. Ann. Oncol 2014; 25: 1656−1663.

75.   Del Conte G, Sessa C, Von Moos R, Viganoo L, Digena T, Locatelli A, Gallerani E, Fasolo A, Tessari A, Cathomas R, Gianni L. Phase I study of olaparib in combination with liposomal doxorubicin in patients with advanced solid tumours. Br. J. Cancer 2014; 111: 651−659.

76.   Liu JF, Tolaney SM, Birrer M, Fleming GF, Buss MK, Dahlberg SE, Lee H, Whalen C, Tyburski K, Winer E, Ivy P, Matulonis UA. A Phase 1 trial of the poly (ADP-ribose) polymerase inhibitor olaparib (AZD2281) in combination with the anti-angiogenic cediranib (AZD2171) in recurrent epithelial ovarian or triple-negative breast cancer. Eur. J. Cancer 2013; 49: 2972−2978.

 

 

 

 

 

 

 

 

Received on 25.01.2019          Modified on 20.02.2019

Accepted on 29.03.2019        © RJPT All right reserved

Research J. Pharm. and Tech. 2019; 12(6):3098-3104.

DOI: 10.5958/0974-360X.2019.00524.9