Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

REVIEW may lead to DNA alterations, including deletions

of genetic material (15–17). Some of the mutations

PARP inhibitors: Synthetic lethality


that arise in this way may foster cancer initiation
or progression, potentially explaining at least
in part why mutations in BRCA1 and BRCA2 in-

in the clinic crease cancer risk; additional roles of BRCA1 and


BRCA2 in processes such as chromatin remodel-
ing and transcriptional regulation may also be
Christopher J. Lord1* and Alan Ashworth2* relevant to pathogenesis (18).
In 2005, two groups described the synthetic
PARP inhibitors (PARPi), a cancer therapy targeting poly(ADP-ribose) polymerase, are the lethal (SL) interaction (Fig. 1A) between PARP
first clinically approved drugs designed to exploit synthetic lethality, a genetic concept inhibition and BRCA1 or BRCA2 mutation, sug-
proposed nearly a century ago. Tumors arising in patients who carry germline mutations gesting a novel strategy for treating patients with
in either BRCA1 or BRCA2 are sensitive to PARPi because they have a specific type of BRCA-mutant tumors (19, 20). SL is a concept
DNA repair defect. PARPi also show promising activity in more common cancers that introduced nearly a century ago by geneticists to
share this repair defect. However, as with other targeted therapies, resistance to PARPi describe the situation whereby a defect in either
arises in advanced disease. In addition, determining the optimal use of PARPi within drug one of two genes has little effect on the cell or
combination approaches has been challenging. Nevertheless, the preclinical discovery organism, but a combination of defects in both
of PARPi synthetic lethality and the route to clinical approval provide interesting lessons genes results in death (21). The demonstration
for the development of other therapies. Here, we discuss current knowledge of PARP that BRCA-mutant tumor cells were as much as
inhibitors and potential ways to maximize their clinical effectiveness. 1000 times more sensitive to PARPi than BRCA-

Downloaded from http://science.sciencemag.org/ on December 25, 2020


wild type cells (depending on the PARPi used and

D
NA damage and its repair or lack thereof ment for most cancer patients. By inhibiting PARP- the experimental format) (19) provided the im-
are central to the induction of mutations, mediated repair of DNA lesions created by chemo- petus for PARPi to be tested in clinical trials as
which drive the development of nearly all or radiotherapy, greater potency might be achieved. single agents. Originally it was proposed that the
cancers. Healthy cells defend themselves About 30 years ago, small-molecule nicotinamide mechanism underlying the SL interaction was
against the deleterious effects of DNA dam- analogs were shown to inhibit PARylation and that PARP inhibition caused persistent SSBs, which,
age through an interrelated series of molecular to enhance the cytotoxicity of dimethyl sulfate, when encountered by a replication fork, some-
pathways, the DNA damage response (DDR), that a DNA damaging agent (7–9). Subsequent drug times resulted in collapse of the fork, potentially
recognize DNA damage, stall the cell cycle, and discovery efforts led to the development of clin- creating a DSB (19). However, this initial model
mediate DNA repair, thus maintaining the in- ical PARPi, including veliparib (Abbvie), rucaparib has recently been modified as a result of data
tegrity of the genome. Key to the DDR are the (Pfizer/Clovis), olaparib (KuDOS/AstraZeneca), and suggesting that some PARPi (especially rucaparib,
poly(ADP-ribose) polymerase 1 and 2 (PARP1 and niraparib (Merck/Tesaro). More recently, a sec- olaparib, niraparib, and talazoparib) “trap” PARP1
PARP2) enzymes, DNA damage sensors and signal ond generation, more potent PARPi, talazoparib on DNA, preventing autoPARylation and PARP1
transducers that operate by synthesizing negatively (Lead/Biomarin/Medivation/Pfizer) has also been release from the site of damage and therefore
charged, branched poly(ADP-ribose) (PAR) chains developed (10). These PARPi all interact with the interfering with the catalytic cycle of PARP1
(PARylation) on target proteins as a form of post- binding site of the PARP enzyme cofactor, b nicotin- (22–24) (Fig. 1, B and D). This trapped PARP1
translational modification (1). PARP1 binds dam- amide adenine dinucleotide (b-NAD+), in the cata- protein has been suggested to be the relevant
aged DNA at single-strand DNA breaks (SSBs) lytic domain of PARP1 and PARP2 but, as discussed cytotoxic lesion at least for some PARPi, a sit-
and other DNA lesions, an event that causes a later, have differing effects in terms of their cytotoxic uation analogous to the mechanism of action
series of allosteric changes in the structure of potency and ability to “trap” PARP1 on DNA. of cancer drugs that inhibit topoisomerase II,
PARP1 that activate its catalytic function (1–5) Carriers of deleterious heterozygous germ- which also “trap” a DNA repair protein on the
(Fig. 1). This leads to the PARylation and recruit- line mutations in the BRCA1 and BRCA2 genes double helix. Supporting this contention, PARP1-
ment of DNA repair effectors such as XRCC1, as have substantially elevated risks of developing defective cells appear to be resistant to PARPi
well as the remodeling of chromatin structure breast, ovarian, and other cancers (11–13). Because (22, 25). Clinically used PARPi differ in their abil-
around damaged DNA as part of the DNA re- the wild-type BRCA allele is lost during tumor- ity to trap PARP1; talazoparib is approximately
pair process. PARP1 eventually PARylates itself igenesis, these genes are considered classical 100 times more potent than niraparib in this
(autoPARylation). The negative charge that PAR tumor suppressors. Both BRCA1 and BRCA2 pro- respect, which in turn traps PARP1 more potently
chains impart upon PARP1 likely causes its release teins are critical to the repair of double-strand than olaparib and rucaparib (23) (Fig. 1C). In con-
from repaired DNA (1–5) (Fig. 1B). DNA breaks (DSBs) by a process called homolo- trast, veliparib appears to have a limited ability to
An understanding of the functions of PARP1 gous recombination repair (HRR), a form of DNA trap PARP1, despite its ability to inhibit PARylation
and PARP2 in the DDR drove long-standing ef- repair that uses a homologous DNA sequence to (Fig. 1C). These differences in PARP1 trapping,
forts to develop small-molecule PARP1/2 inhib- guide repair at the DSB. HRR is generally a “con- rather than simply the ability to inhibit PARylation,
itors (PARPi) (Fig. 1C) (6). The original rationale servative” mechanism, in that it restores the may be a better predictor of in vitro cytotoxicity
was that PARPi could sensitize tumor cells to original DNA sequence at the site of DNA damage in BRCA-mutant cells, with talazoparib having
conventional treatments that cause DNA damage, (14). When cells become HRR deficient, whether the most profound cytotoxic effects (10, 22, 23).
including multiple chemotherapy or radiotherapy driven by defects in BRCA1, BRCA2, or other Differences in PARP1 trapping activity may also
approaches, which remain the backbone of treat- pathway components, nonconservative forms of need to be a consideration when designing com-
DNA repair predominate, such as nonhomolo- bination therapies involving PARPi (see below);
gous end joining (NHEJ). These processes either it is possible that the ability to trap PARP1 may
1
The Cancer Research UK Gene Function Laboratory and
fuse broken DNA ends at the DSBs without using produce unacceptable toxicity when some PARPi
Breast Cancer Now Toby Robins Research Centre, The a homologous DNA sequence to guide repair or are combined with conventional doses of cyto-
Institute of Cancer Research, London SW3 6JB, UK. fuse regions of DNA close to the site of the DSB toxic chemotherapies. PARP1 and PARP2 have
2
University of California, San Francisco (UCSF), Helen Diller that exhibit short regions of DNA sequence homol- multiple important roles beyond the DDR, such as
Family Comprehensive Cancer Center, 1450 Third Street, San
Francisco, CA 94158, USA.
ogy, deleting the intervening DNA sequence. The transcription, apoptosis, and immune function;
*Corresponding author. Email: chris.lord@icr.ac.uk (C.J.L.); preferential use of these nonconservative repair the antitumor efficacy of PARPi might also re-
alan.ashworth@ucsf.edu (A.A.) mechanisms in the absence of HRR therefore flect alterations in these functions (3).

Lord et al., Science 355, 1152–1158 (2017) 17 March 2017 1 of 6


F R ON T IER S I N C A N CE R TH E R AP Y

A B i ZnF
WGR HD ART catalytic
BRCT domain
2
Protein A Protein B 1 3
Cell survives
No alterations C

ii Auto-Parylation vi
Cell survives
Protein A altered “Trapping”
Single-strand PARP inhibitor
break in DNA vii
Cell survives PARP trapped
on DNA: blocks
Protein B altered
replication fork
progression,
requires HRR PARP1
Cell death substrate
for repair.
protein
Both altered

v
iii
2 1 3 β-NAD+

iv PARylation via
C

Downloaded from http://science.sciencemag.org/ on December 25, 2020


an active ART
PARP trapping potency (high to low) catalytic domain

1
2 3, 4
5
H
O NN
NN O NH2 D
R
N N NH i Direction of travel v PARP inhibitor resistance and
F N tumor cell survival caused by
NS of replication fork
H multiple distinct mechanisms
F
1 Talazoparib 2 Niraparib
H H H O iii DNA repair
N N F O NN F by HRR,
N Tumor cell • Secondary “reversion”
N mutations in BRCA1, BRCA2,
survival
O O RAD51C/D
N
3 Rucaparib H 4 Olaparib ii iv Defects in HRR = • Restoration of HRR in
Stalled synthetic BRCA1 mutant tumor cells via
O NH2 replication fork lethality and loss of 53BP1, REV7
tumor cell death • Loss of PARP1 expression
N
N N • Pharmacological resistance
H H e.g. upregulation of
5 Veliparib P-glycoprotein pumps

Fig. 1. Mechanism of action of PARPi. (A) Schematic of synthetic lethality. In target protein), mediating the recruitment of DNA repair effectors, chromatin
its simplest form, the simultaneous alteration of two genes or proteins (shown remodeling, and eventually DNA repair. (vi) PARP1 autoPARylation (likely in cis
here as A and B) causes cell death, while alteration of either gene/protein alone at SSBs but possibly in trans at other DNA lesions (4) finally causes the release
does not. When the concept is applied to cancer treatment, where gene A rep- of PARP1 from DNA and the restoration of a catalytically inactive state [shown
resents an oncogene, tumor suppressor gene, or oncogenic process/pathway, in (i)]. (vii) Several clinical PARPi, each of which binds the catalytic site, prevent
gene B, once identified, becomes a candidate therapeutic target that can be the release of PARP1 from DNA, “trapping” PARP1 at the site of damage, po-
used to target tumor cells with dysfunction in A. (B) A model describing the tentially removing PARP1 from its normal catalytic cycle. These images are
PARP1 catalytic cycle. (i) In its non-DNA bound state, PARP1 exists in a rela- schematic; detailed structures and models of PARP1/DNA nucleoprotein com-
tively disordered conformation, commonly referred to as “beads on a string” (4). plexes are described elsewhere (4, 5). (C) Clinical PARP inhibitors. Chemical
The domain structure of PARP1 is shown, including three zinc finger–related structures of five clinical PARPi are shown. The ability of each PARPi to trap
domains (ZnF 1, 2, and 3): the BRCA1 C-terminus domain (BRCT); the PARP1 on DNA differs (talazoparib being the most potent PARP1 trapping
tryptophan-, glycine-, arginine-rich domain (WGR); and the catalytic domain, inhibitor, veliparib being the least potent) and broadly correlates with cytotoxic
which encompasses two subdomains; a helical domain (HD) and an ADP- potency (22–24). (D) A model of PARP inhibitor synthetic lethality. Trapped
ribosyltransferase (ART) catalytic domain. In this non-DNA bound state, HD PARP1/DNA nucleoprotein complexes impair the progression of replication
acts as an autoinhibitory domain preventing binding of the PARP-superfamily forks. (i) Schematic of trapped PARP1 on DNA in front of a replication fork;
GRAPHIC: ADAPTED BY V. ALTOUNIAN/SCIENCE

cofactor, b-NAD+, to its ART binding site (5). (ii) Damage of the DNA double newly synthesized DNA is shown in red. (ii) The replication fork is impeded by
helix often causes the formation of SSBs (predamaged and damaged DNA trapped PARP1. This normally induces a DNA damage response. (iii) HRR,
structures are shown); SSBs cause a change in the normal orientation of the involving BRCA1 and BRCA2 tumor-suppressor proteins, is the optimal DNA
double helix, which, in turn, (iii) provides a binding site for DNA binding PARP1 repair process for repairing and restarting replication forks stalled by PARPi
ZnF domains. The interaction of ZnF 1, 2, and 3 with DNA initiates a stepwise and also involves the use of additional “BRCAness” proteins. In the absence of
assembly of the remaining PARP1 protein domains onto the PARP1/DNA nu- effective HRR, cells use DNA repair processes that can potentially generate
cleoprotein structure, shown in (iv); this process leads to a change in HD large-scale genomic rearrangements, which often leads to tumor cell death
conformation, and resultant loss of autoinhibitory function, thus allosterically and synthetic lethality. (v) Even where HRR is defective, PARPi resistance oc-
activating PARP1 catalytic activity (5). (v) ART catalytic activity drives the curs. Multiple mechanisms cause PARPi resistance but can be broadly clas-
PARylation of PARP1 substrate proteins (branched PAR chains are shown on a sified into the examples shown.

Lord et al., Science 355, 1152–1158 (2017) 17 March 2017 2 of 6


As with other targeted therapies, acquired re- and subsequent PARPi responses (36). Using response; this observation was consistent with
sistance to PARPi therapy has been observed in mutational signatures (i.e., the number and the hypothesis that platinum-based drugs and
most patients with advanced cancer (Fig. 1D). type of mutations found in a tumor) to identify PARPi target similar molecular defects (36). Phase
Multiple potential mechanisms of resistance to BRCAness is based on the observation that the 2 trials involving patients with gBRCAm breast,
PARPi have been identified through in vitro ex- tumor genomes of cancer patients with germline ovarian, pancreatic, or prostate cancers confirmed
perimentation. These include inactivation of the BRCA gene mutations (gBRCAm) tend to display that olaparib offered clinical benefit (46–48)
DNA repair proteins 53BP1 (26) or REV7 (27), a characteristic pattern of mutations—a muta- (Fig. 2B). The response rate in gBRCAm ovarian
which results in the restoration of HRR. Loss of tional “scar” that includes large-scale genomic cancer patients (48) was sufficient for the U.S.
a number of proteins, including PARP1 itself (25), rearrangements—that likely reflects the prefer- Food and Drug Administration (FDA) to ap-
which are involved in maintaining replication ential use of nonconservative DNA repair mech- prove olaparib as a treatment for patients with
fork stability, is also proposed to cause PARPi anisms in the absence of HRR over the lifetime advanced ovarian cancer who had already re-
resistance (28). Secondary “revertant” mutations of the disease; similar mutational scars are also ceived three or more previous lines of therapy
in BRCA1 or BRCA2 that restore the open read- apparent in tumors that do not have BRCA gene (49) (Fig. 2B).
ing frame of the genes and restore sufficient HRR mutations, raising the possibility that the pres- A number of studies sought to extend these prom-
function also lead to PARPi resistance (29, 30); ence of such a “BRCAness scar” could be used ising results to patients with BRCAness tumors:
these secondary mutations also cause clinical resist- to predict clinical responses to agents such as high-grade serous ovarian cancers (HGSOvCa),
ance to platinum-based chemotherapy (29, 31–34). PARPi or platinum-based drugs in patients without triple negative breast cancers (TNBC), and ad-
The latter mechanism is the only one so far to be gBRCAm (34). Which of these molecular profiling– vanced prostate cancers. In HGSOvCa, olaparib
fully clinically validated as a mechanism of re- based biomarkers will be most effective in predict- was found to reduce the rate of disease recur-
sistance to PARPi. As with other targeted ther- ing clinical responses to drugs such as PARPi, rence and extend progression-free survival when
apies, the selective pressure that PARPi provide however, is not yet clear. For example, although used as a maintenance therapy in patients pre-

Downloaded from http://science.sciencemag.org/ on December 25, 2020


in BRCA-defective tumor cells drives the emer- mutational scars of BRCAness, as they are cur- viously treated with platinum-based chemothera-
gence of resistant clones, at least in advanced rently measured, probably reflect the mutational py (50–52). These observations led to the approval
cancers. Alternative treatment strategies that sup- processes that alter the genome in the absence of of olaparib by the European Medicines Agency
press or at least delay the emergence of resistant HRR over the entire lifetime of a tumor, they might (EMA) as a maintenance treatment for BRCA-
clones are therefore now required. Moreover, not provide an accurate estimate of whether mutant patients with platinum-sensitive gyne-
as some of these mechanisms cause resistance HRR is still defective in tumor cells at the time cological cancers (high-grade serous epithelial
to both PARPi and platinum-based drugs, careful that treatment is delivered, a likely prerequisite ovarian, fallopian tube, or primary peritoneal
consideration needs to be given to the therapies for a profound and sustained antitumor response cancers). In this particular case, olaparib was ap-
provided to patients both before and after PARPi to a PARPi, or even how plastic or reversible the proved for use in patients with either germline or
treatment. extant HRR defect is. Although a further refine- somatically occurring BRCA mutations (gBRCAm
The term “BRCAness” has been used to de- ment of how such scars are quantified might or sBRCAm, respectively) (Fig. 2B). BRCAness-
scribe tumors that have not arisen from a germ- allow more accurate predictions to be made (for related clinical benefit has also been seen in meta-
line BRCA1 or BRCA2 mutation (gBRCAm) but example, by assessing the clonality of BRCAness static, castration-resistant prostate cancer: In the
nonetheless share certain phenotypes, in par- mutational scars across multiple clones within a TO-PARP phase 2 clinical trial, about 30% of pa-
ticular an HRR defect, with these hereditary can- single tumor), one solution to this problem might tients showed a clinical response to olaparib,
cers (34, 35). For example, somatically occurring be to directly measure HRR functionally in tumor half of whom had BRCA2 or ATM defects in their
mutations in either BRCA1 or BRCA2 at least par- specimens using approaches such as the localiza- tumors (39). In TNBC, clinical responses to ola-
tially phenocopy gBRCAm in terms of causing tion or activity of key DNA repair proteins that parib were somewhat mixed; although patients
an HRR defect, as does somatic promoter hyper- are involved in this DNA repair process, such as with BRCA1/2-mutant tumors showed some dis-
methylation of BRCA1 or mutations in other genes RAD51 (34, 42, 43) (Fig. 2A). Although this has ease stabilization when treated with olaparib,
that are involved in DSB repair and the stability not yet been tested, the use of functional HRR there were no sustained responses in either BRCA1/
of replication forks (34, 35). Consistent with the biomarkers alongside BRCAness scar assessments 2-mutant or nonmutant cohorts, suggesting that
HRR defect, tumors with BRCAness might also might turn out to be the most effective way of BRCAness might be less frequent in TNBC than
share therapeutic vulnerabilities with gBRCAm identifying BRCAness. in diseases such as HGSOvCa (53) or that other
tumors, such as sensitivity to platinum-based drugs clinical parameters or previous treatment effects
(36). Shortly after the demonstration that BRCA1- The clinical development of PARPi might be important.
or BRCA2-mutant cells were highly susceptible PARPi first entered human clinical trials as a The recent approval of olaparib for ovarian
to PARPi, deficiencies in a number of tumor sup- combination therapy consisting of low-dose ru- cancer was followed by promising phase 3 clini-
pressor genes involved in HRR, such as ATM, caparib and full-dose temozolomide, a DNA alkyl- cal results with niraparib in ovarian cancer pa-
ATR, PALB2, and the FANC gene family, were ating agent (44). Based on the preclinical data tients as a maintenance therapy (54). Patients
also shown to confer sensitivity to PARPi (34, 37). showing an SL interaction between PARP inhibi- with platinum-sensitive, recurrent ovarian cancer
Large-scale cancer genome–sequencing projects tion and BRCA mutation status (19, 20), a phase were categorized according to the presence or
also revealed that somatic mutations in genes 1 clinical trial of olaparib, including patients with absence of gBRCAm. Patients without a gBRCA
involved in HRR occur in a wide spectrum of germline BRCA1 or BRCA2 mutations (gBRCAm), mutation (non-gBRCAm) were further character-
tumors (34), in particular in substantial fractions was initiated. In this trial, 63% of the gBRCAm- ized to identify those without a gBRCAm whose
of high-grade serous ovarian cancer (HGS-OVCa) mutant patients demonstrated a clinical benefit, tumors nevertheless exhibited a mutational pat-
(38), advanced prostate cancer (39), and pancre- thus confirming the SL hypothesis (45). Dose- tern often seen in the genome of gBRCAm tumors—
atic cancer (40, 41). These and other cancers with limiting myelosuppression and central nervous a BRCAness DNA scar, described above (55). Those
HRR mutations are therefore candidates for test- system side effects were observed in some pa- patients who received niraparib showed longer
ing PARPi efficacy. tients, but overall, side effects were less severe progression-free survival (PFS) than those who
Considerable effort has also been invested in than those seen with conventional chemotherapy received placebo, with the strongest effects being
other molecular profiling approaches, such as regimens (45). Expansion of this trial to 50 gBRCAm seen in the gBRCAm group. In the non-gBRCAm
transcriptional and mutational signature profiling, patients with gynecological malignancies con- cohort, niraparib also extended PFS, especially in
that might identify BRCAness and PARPi-sensitive firmed efficacy and demonstrated a correlation patients whose tumors had somatic BRCA muta-
tumors (34), as well as considering previous plat- between a favorable response to previous plat- tions (sBRCAm), who displayed a similar reduc-
inum sensitivity as a predictor of BRCAness inum chemotherapy and subsequent olaparib tion in the risk of disease progression as those

Lord et al., Science 355, 1152–1158 (2017) 17 March 2017 3 of 6


F R ON T IER S I N C A N CE R TH E R AP Y

with gBRCAm. Overall, BRCAness-scar–positive


patients displayed longer PFS compared with Predictive biomarkers of PARP inhibitor sensitivity.
BRCAness-scar–negative patients, although the ulti-
mate predictive value of this scar biomarker still FDA approved,
requires clarification in larger phase 3 trials (54, 56). Experimental Examples
EMA approved
Recently, rucaparib has also been shown to Somatic, deleterious DNA sequencing of
extend PFS in a maintenance setting (57). In the mutations in other individual genes implicated
Germ-line BRCAness genes known in HRR or “panel”
ARIEL2 phase 2 trial (NCT01891344), patients
deleterious to modulate HRR and sequencing of multiple
with platinum-sensitive, high-grade ovarian carci- mutations genes
Liquid biopsy other DNA repair
noma were classified into three groups according in either processess.
to the genomic features of their tumors, including BRCA1 or BRCA2 Detection of “revertant”
an assessment of a proposed BRCAness-related mutations that cause
DNA mutational scar assessed by calculating the PARPi resistance
Somatic, Tumor biopsy
extent of chromosomal loss of heterozygosity (LOH)
deleterious
across the genome: (i) BRCA mutant (gBRCAm Transcriptomic
mutations
or sBRCAm); (ii) BRCA wild-type but with high in either signatures
genomic LOH; and (iii) BRCA wild-type with low BRCA1 or BRCA2
LOH. PFS was greatest for the BRCA-mutant group, Functional biomarkers Immunohistochemical
with the high-LOH group displaying slightly more of HRR detection of DNA repair
proteins involved in HRR
benefit from rucaparib treatment than in the
low-LOH group (57). However, the fraction of pa- Genomic “scar” Use of deep sequencing or

Downloaded from http://science.sciencemag.org/ on December 25, 2020


tients exhibiting durable responses in these BRCA signatures DNA copy number analysis
to estimate the presence
wild-type groups was still below that expected of chromosomal
with standard platinum-based combination chemo- rearrangements similar
therapies, suggesting that further studies are re- to those found in gBRCAm
quired to justify the use of PARPi in these BRCA cancers
wild-type patients, as opposed to platinum-based
treatments (56, 57). A subsequent phase 3 clinical Clinical assessment of PARPi synthetic lethality.
study assessing rucaparib (ARIEL 3, NCT01968213)
will no doubt clarify this issue and also confirm
whether the distinction in clinical responses be-
• Breast cancer Substantial fractions of BRCA1, BRCA2 mutations and
BRCAness alterations
tween LOH high and low groups can be replicated
and whether refined criteria for converting a • Pancreatic cancer Phase 2 and 3 clinical trials on-going
genome-wide loss of heterozygosity score into a Olaparib has FDA breakthrough therapy designation in
clinically applicable BRCAness biomarker can be • Prostate cancer metasatic, castration-resistant prostate cancer
achieved (56). Nevertheless, these clinical results,
when taken together with additional phase 2 data • Ovarian cancer
in HGSOvCa [NCT01482715 (58)], have led to FDA Substantial fractions of BRCA1, BRCA2 mutations and BRCAness alterations
approval for rucaparib for use in patients with Clinical trials ongoing. Approvals include:
gBRCAm or sBRCAm advanced ovarian cancer EMA approval for olaparib (2014) monotherapy for the maintenance treatment of adult
who have been treated with two or more chemo- patients with platinum-sensitive, relapsed, BRCA-mutated (germline and /or somatic)
therapies (Fig. 2B). high-grade serous epithelial ovarian, fallopian tube, or primary peritoneal cancer who are in
Talazoparib is also starting to show clinical response (complete response or partial response) to platinum-based chemotherapy
potential. In a recent study, 13 early-stage breast
cancer patients with germline mutations in either FDA approval for olaparib (2014) monotherapy in patients with deleterious or suspected
BRCA1 or BRCA2 were treated for two months germline BRCA-mutated advanced ovarian cancer detected by FDA-approved test who have
with talazoparib in a “neoadjuvant” setting—i.e., been treated with >
_ 3 prior lines of chemotherapy
before chemotherapy and surgery. All patients
FDA approval for rucaparib (2016) monotherapy in patients with deleterious BRCA
displayed a reduction in tumor volume after mutation (germline and/or somatic)-associated advanced ovarian cancer who have been
2 months (59). This study is now being expanded treated with >
_ 2 prior lines of chemotherapy
to assess the effects of 4 to 6 months of neoad-
juvant talazoparib therapy. Similar neoadjuvant
studies assessing rucaparib in breast cancer are Fig. 2. Clinical PARPi synthetic lethality. (Top) Predictive biomarkers of PARP inhibitor sensitivity. Com-
also under way. panion FDA- and EMA-approved tests, which detect the presence of either germline or somatic BRCA1 or
It would be premature to draw conclusions BRCA2 mutations, are currently used to identify patients likely to respond to PARPi therapy. Experimental
about which PARPi are most effective in particu- biomarkers are nonapproved biomarkers where the sensitivity (true positive rate/proportion of positives
lar patient subsets; at present, the different clinical that are correctly identified) and/or specificity (true negative rate) are not yet clear. These include ap-
trial designs and distinct patient populations used proaches that estimate the presence/absence of an HRR defect via the identification of DNA mutations in
GRAPHIC: ADAPTED BY N. CARY/SCIENCE

to assess different PARPi preclude such direct BRCAness genes that control tumor cell responses to PARPi, approaches that estimate an HRR defect by
comparisons. However, a burgeoning dissection identifying the extent and type of chromosomal alterations often found in BRCA–mutant and BRCAness
of the biochemical and cellular effects of differ- tumors, and also functional biomarkers that use the visualization of key proteins involved in HRR as a
ent PARPi is beginning to influence ideas on how predictor of the ability to repair PARPi-induced DNA lesions. (Bottom) Clinical assessment of PARPi syn-
different PARPi might be used clinically. For thetic lethality. Most clinical trials assessing PARPi synthetic lethality have focused on tumor types that
example, although talazoparib can kill BRCA- exhibit subsets of either germline BRCA1 or BRCA2 mutations or other candidate BRCAness defects.
mutant cells in vitro at a dose lower by a factor of Regulatory bodies including the FDA and EMA have recently approved PARPi to be used in ovarian cancer
200 than the dose needed for olaparib and ruca- patients with either BRCA1 or BRCA2 mutations (as shown), with these being detected via companion
parib [an effect that correlates with the superior diagnostic assays.

Lord et al., Science 355, 1152–1158 (2017) 17 March 2017 4 of 6


PARP1 trapping activity of talazoparib (23)], the of similar “high PARPi/low chemo” approaches resistance could perhaps be mitigated with two
in vitro therapeutic ratio achieved in BRCA1/2- (e.g., NCT02049593; ClinicalTrials.gov). In addi- orthogonal treatments.
defective cells versus wild-type cells is similar for tion, preclinical evidence suggests that a series of In addition to identifying well-tolerated PARPi
all three PARPi (10). It is therefore unclear wheth- targeted agents, such as inhibitors of the phos- combination therapies, defining predictive bio-
er the enhanced trapping ability of talazoparib phatidylinositol 3-kinase signaling pathway, can markers that facilitate patient stratification for
compared with rucaparib and olaparib will in it- impart or enhance a BRCAness phenotype in such combinations is a key objective, as is iden-
self lead to an improved therapeutic ratio (10). tumor cells, thus causing sensitivity to PARPi (63); tifying combination therapies that target PARPi
Furthermore, mass spectrometry–based analysis clinical studies testing the applicability of this resistance mechanisms and therefore provide
of PARPi binding proteins suggests that different approach are also now under way. In a distinct more durable clinical responses. Finally, to deliv-
PARPi have distinct interaction profiles (60). How- strategy, the concept of combining PARPi with er the best overall response, the scheduling of
ever, it is not clear how or whether differences in targeted agents that impair the ability of tumor PARPi and its sequencing with other drugs in the
the “off-target” profile might alter the antitumor cells to stall the cell cycle to process and repair combination should be given greater attention.
effects of PARPi or contribute to adverse side ef- “trapped” PARP1 DNA lesions (e.g., WEE1, ATR,
fects. An understanding of which PARPi is the pre- and CHEK1/2 inhibitors) has gained some trac- Conclusions and future prospects
ferred agent in a given clinical situation will likely tion of late, being bolstered by the recent deve- The successful development of PARPi for BRCA-
be informed by a future comparison of mature lopment of druglike WEE1 and ATR inhibitors mutant cancers provides proof of concept that
phase 3 clinical outcome data. that now make this a testable hypothesis in SL interactions can be translated into cancer
therapies. Although it remains to be seen how
many other SL interactions can be clinically ex-
ploited, a number of lessons can be learned from
the discovery and development of the PARPi/

Downloaded from http://science.sciencemag.org/ on December 25, 2020


Box 1. Some key unanswered questions about PARP inhibitors.
BRCA SL interaction. For example, SL interac-
• What proteins beyond BRCA1 and BRCA2 contribute to the processing of trapped PARP1, the tions with therapeutic value are ideally (i) as-
drug’s key cytotoxic DNA lesion? sociated with a therapeutic window defined by a
• How does the inability to repair a trapped PARP1 lesion at the replication fork translate into biomarker that can be used to stratify patients
cell death? for therapy; (ii) capable of highly penetrant ef-
• How do the roles of PARP1 in processes unrelated to DNA repair (e.g., inflammation, apoptosis, fects, where the presence of the biomarker pre-
immune system) influence the anticancer activity of PARPi? dicts profound sensitivity to inhibition of the
• What is the relative predictive value of BRCA1, BRCA2 gene mutations or BRCAness biomarkers SL target in the majority of cases; (iii) robust in
for platinum and/or PARPi responses. What is the best way to measure BRCAness? the face of the molecular diversity and plasticity
• What mechanisms distinguish “superresponders,” who show profound and sustained responses seen in human tumors; (iv) pharmacologically
to PARPi, from those who do not? tractable; and (v) well understood in terms of
• What mechanisms operate clinically to drive development of resistance to PARPi in both molecular mechanism, as this can inform the
BRCA-mutant cancers and BRCAness cancers? development of biomarkers and an understand-
• How can PARPi be optimally used in combination therapies? ing of drug resistance mechanisms. In the case of
PARPi, the scale of the therapeutic effect in in vitro
preclinical models with BRCA mutations was
compelling enough to drive early entry of the
Potential PARPi combination therapies clinical trials [e.g., NCT02264678, NCT02576444 drugs into hypothesis-testing clinical trials. An
As discussed above, the ability of PARPi to sensi- (62)]. Finally, combinations of PARPi with im- early assessment of the robustness and penetrance
tize tumor cells to DNA-damaging chemothera- munotherapies such as antibodies to CTLA4 and of the BRCA/PARPi SL interactions indicated that
pies provided the initial rationale for developing PD1/PDL-1 are now being tested clinically; these these were highly penetrant and relatively re-
clinically useful PARPi drugs. PARPi sensitize cells studies are based partly on the hypothesis that sistant to genetic background effects and addi-
to the alkylating agent temozolomide as well as tumors with BRCA1, BRCA2, or BRCAness defects tional molecular alterations. The use of preclinical
to topoisomerase poisons and inhibitors, effects have a higher mutagenic burden and therefore approaches to identify mechanisms of drug re-
that are now becoming mechanistically under- potentially an elevated neo-antigen load, which sistance alongside early-stage clinical trials pro-
stood. Temozolomide sensitization is caused by is thought to produce a stronger antitumor im- vided further insights into the mechanism of
PARP1 trapping, whereas topoisomerase poison mune response (64–65). In addition, there is action of PARPi and revealed the basis of some
sensitivity is largely driven by the catalytic inhibi- evidence that BRCA deficiency may induce a of the incomplete clinical responses to PARPi
tion of PARP1 and less dependent upon trapping STING-dependent innate immune response (66), in clinical trials (29, 30). Finally, a critical factor
ability (22, 23). These observations are broadly which might also influence the antitumor effect of in the rapid translation to the clinic of the SL
consistent with the failure of the relatively poor PARPi/immunotherapy drug combinations. In concept was the availability of potent drug-like
PARP1 trapping PARPi, veliparib, to enhance clin- this regard, trials such as MEDIOLA (NCT02734004), PARPi at the same time as the identification of
ical temozolomide responses in BRCA gene mu- a phase 1/2 trial of olaparib in combination with the BRCA SL (19, 20), highlighting the impor-
tant breast cancer patients, despite enhancing the durvalumab (MEDI4736), an anti–PD-L1 immune tance of medicinal chemistry and pharmacology
effect of a carboplatin/paclitaxel combination checkpoint inhibitor, may be informative. The ra- to the successful application of PARP/BRCA SL
(61). However, clinical experience with therapies tionale for the identification of efficacious PARPi and exemplifying how the convergence of other-
that combine PARPi with chemotherapies has combinations has focused largely on enhancing wise distinct disciplines such as cancer genetics
been, in general, mixed. Dose-limiting normal the antitumor effect of PARPi by creating DNA and drug discovery can be effective.
tissue toxicity is frequently observed, especially damage or modulating DNA repair. However, Ideally, the design and interpretation of clinical
when the commonly used dose of the chemother- PARPi could also be beneficially combined with trials based on SL interactions should be based
apy regimen is combined with a PARPi (62). As drugs that act by targeting cancer-specific fea- on the biological hypothesis and robust preclinical
some preclinical studies have confirmed that tures that are superficially unrelated to the DDR data. Although there is a direct link between the
tumor cell growth inhibition can be achieved or BRCA function; the promising combination of recent approvals for PARPi in BRCA1/2-mutant
using high-dose PARPi combined with relative- olaparib with the antiangiogenic agent cedir- cancers and the original preclinical data identi-
ly low doses of chemotherapy, ongoing clinical anib is an example of this (67, 68). One poten- fying the SL interactions over a decade ago, the
studies are assessing the tolerability and efficacy tial advantage of such an approach is that therapy clinical development of PARPi as a SL approach

Lord et al., Science 355, 1152–1158 (2017) 17 March 2017 5 of 6


F R ON T IER S I N C A N CE R TH E R AP Y

has not been straightforward. For example, a tiple cancers. We suggest three broad areas, the 35. N. Turner, A. Tutt, A. Ashworth, Nat. Rev. Cancer 4, 814–819
purported PARPi, iniparib, failed to elicit the “holy trinity” of personalized cancer therapy re- (2004).
36. P. C. Fong et al., J. Clin. Oncol. 28, 2512–2519 (2010).
expected clinical responses in a phase 3 trial, search, that require further investigation: (i) Iden-
37. N. McCabe et al., Cancer Res. 66, 8109–8115 (2006).
despite showing potential in early-stage clinical tifying who to treat. This can be achieved by 38. D. Bell et al., Nature 474, 609–615 (2011).
assessment (69). As a result, questions were raised dissecting the mechanisms by which PARPi kill 39. J. Mateo et al., N. Engl. J. Med. 373, 1697–1708 (2015).
about the clinical potential of the entire drug class or inhibit tumor cells and using this information 40. N. Waddell et al., Nature 518, 495–501 (2015).
and the SL approach in general (69). In retrospect, to develop refined, mechanism-based, biomarkers 41. J. Carnevale, A. Ashworth, J. Clin. Oncol. 33, 3080–3081
(2015).
the evidence supporting the mechanism of action that allow patient stratification. (ii) Combating 42. M. Graeser et al., Clin. Cancer Res. 16, 6159–6168 (2010).
of iniparib as a PARPi was not compelling (69). drug resistance. This can be achieved by identify- 43. A. Mukhopadhyay et al., Clin. Cancer Res. 16, 2344–2351
This reinforces the argument for the clinical de- ing the mechanisms that cause PARPi resistance (2010).
velopment of drugs to be informed by robust pre- and biomarkers that predict it, by understanding 44. R. Plummer et al., Cancer Chemother. Pharmacol. 71, 1191–1199
(2013).
clinical biology. Of course, the preclinical and how cancer heterogeneity and plasticity influ-
45. P. C. Fong et al., N. Engl. J. Med. 361, 123–134 (2009).
clinical investigation of PARPi SL effect is far from ence these processes, and by identifying clinical 46. M. W. Audeh et al., Lancet 376, 245–251 (2010).
complete, and we highlight in Box 1 a series of approaches to delay or prevent the emergence 47. A. Tutt et al., Lancet 376, 235–244 (2010).
unanswered questions that, once addressed, could of the drug-resistant phenotype. (iii) Optimizing 48. B. Kaufman et al., J. Clin. Oncol. 33, 244–250 (2015).
guide the optimal use of PARPi in the future. For combination therapy. This can be achieved by 49. G. Kim et al., Clin. Cancer Res. 21, 4257–4261 (2015).
50. J. Ledermann et al., N. Engl. J. Med. 366, 1382–1392
example, a key observation has been that a fraction understanding the mechanistic basis of why some
(2012).
(~15%) of ovarian cancer patients with BRCA1- or drug combinations have synergistic antitumor 51. J. Ledermann et al., Lancet Oncol. 15, 852–861 (2014).
BRCA2-mutant tumors continue to be disease free effects, determining how drug combinations can 52. J. A. Ledermann et al., Lancet Oncol. 17, 1579–1589
more than 5 years after the initiation of PARPi be used to target mechanisms of drug resistance, (2016).
treatment (52). Understanding the underlying and identifying predictive biomarkers of not only 53. K. A. Gelmon et al., Lancet Oncol. 12, 852–861 (2011).

Downloaded from http://science.sciencemag.org/ on December 25, 2020


54. M. R. Mirza et al., N. Engl. J. Med. 375, 2154–2164
reasons for these extended responses could help single agent PARPi responses but also PARPi com- (2016).
in the design of both predictive markers and bination therapy responses. 55. M. L. Telli et al., Clin. Cancer Res. 22, 3764–3773
therapeutic strategies. If these issues can be addressed, we believe that (2016).
Due to advances in technology, the system- PARPi could eventually deliver considerable bene- 56. A. González Martín, Lancet Oncol. 18, 8–9 (2017).
57. E. M. Swisher et al., Lancet Oncol. 18, 75–87 (2017).
atic genome-wide identification of new SL inter- fit to a substantial subset of cancer patients. 58. R. Shapira-Frommer et al., Ann. Oncol. 33 (suppl.), abstr. 2476
actions has now been achieved in yeast (70), and (2015).
mapping each of the SL vulnerabilities asso- RE FERENCES AND NOTES 59. J. K. Litton et al., Ann. Oncol. 27 (suppl. 6), vi43–vi67
ciated with cancer driver genes and oncogenic (2016).
1. M. S. Satoh, T. Lindahl, Nature 356, 356–358 (1992).
60. C. E. Knezevic et al., Cell Chem. Biol. 23, 1490–1503
processes in human cells is an ongoing activity; 2. M. De Vos, V. Schreiber, F. Dantzer, Biochem. Pharmacol. 84,
(2016).
this raises the possibility that additional cancer- 137–146 (2012).
61. S. J. Isakoff et al., Future Oncol. 13, 307–320 (2017).
3. R. Krishnakumar, W. L. Kraus, Mol. Cell 39, 8–24
related SL interactions might be available for (2010).
62. A. Dréan, C. J. Lord, A. Ashworth, Crit. Rev. Oncol. Hematol.
therapeutic exploitation. Critical to these efforts 108, 73–85 (2016).
4. S. Eustermann et al., Mol. Cell 60, 742–754 (2015).
63. Y. H. Ibrahim et al., Cancer Discov. 2, 1036–1047 (2012).
will be a greater understanding of the underly- 5. J. M. Dawicki-McKenna et al., Mol. Cell 60, 755–768
64. T. Higuchi et al., Cancer Immunol. Res. 3, 1257–1268
ing principles of what triggers an SL interaction, (2015).
(2015).
6. T. Zaremba, N. J. Curtin, Anticancer. Agents Med. Chem. 7,
the factors determining the robustness of such 515–523 (2007).
65. J. Huang et al., Biochem. Biophys. Res. Commun. 463, 551–556
interactions (i.e., how easily are SL interactions (2015).
7. S. Shall, J. Biochem. 77 (supp1), 2 (1975)
reversed by other molecular changes), and how 8. M. R. Purnell, W. J. Whish, Biochem. J. 185, 775–777 66. E. E. Parkes et al., J. Natl. Cancer Inst. 109, djw199
(1980). (2016).
robust SL interactions can be predicted, rather 67. S. P. Ivy, J. F. Liu, J. M. Lee, U. A. Matulonis, E. C. Kohn,
9. M. Terada, H. Fujiki, P. A. Marks, T. Sugimura, Proc. Natl. Acad.
than only empirically identified through large- Sci. U.S.A. 76, 6411–6414 (1979). Expert Opin. Investig. Drugs 25, 597–611 (2016).
scale genetic screens. For example, it has been 10. Y. Shen et al., Clin. Cancer Res. 19, 5003–5015 (2013). 68. J. F. Liu et al., Lancet Oncol. 15, 1207–1214 (2014).
suggested that robust SL interactions are enriched 11. M. C. King, Science 343, 1462–1465 (2014). 69. J. Mateo, M. Ong, D. S. Tan, M. A. Gonzalez, J. S. de Bono,
12. Y. Miki et al., Science 266, 66–71 (1994). Nat. Rev. Clin. Oncol. 10, 688–696 (2013).
for pairs of genes that are closely connected on
13. R. Wooster et al., Nature 378, 789–792 (1995). 70. A. H. Tong, C. Boone, Methods Mol. Biol. 313, 171–192
protein-protein interaction networks; i.e., those 14. M. E. Moynahan, M. Jasin, Nat. Rev. Mol. Cell Biol. 11, 196–207 (2006).
that directly interact or interact via one or two (2010). 71. J. Campbell et al., Cell Reports 14, 2490–2501 (2016).
additional proteins or nodes, rather than being 15. M. E. Moynahan, T. Y. Cui, M. Jasin, Cancer Res. 61, 72. J. H. Young, E. M. Marcotte, G3 7, 617–624 (2017).
distantly connected via a larger number of in- 4842–4850 (2001).
16. M. E. Moynahan, A. J. Pierce, M. Jasin, Mol. Cell 7, 263–272 AC KNOWL ED GME NTS
tervening nodes (71). Likewise, proteins involved (2001). C.J.L. acknowledges funding from Breast Cancer Now, Cancer
in similar functions (often predicted from over- 17. A. Tutt et al., EMBO J. 20, 4704–4716 (2001). Research UK, and National Health Service funding to the National
lapping protein-protein interaction networks) are 18. K. K. Dhillon, I. Bajrami, T. Taniguchi, C. J. Lord, Endocr. Relat. Institute for Health Research Royal Marsden Hospital Biomedical
hypothesized to have some shared SL inter- Cancer 23, T39–T55 (2016). Research Centre. A.A. is funded by Susan G. Komen for the Cure,
19. H. Farmer et al., Nature 434, 917–921 (2005). The Breast Cancer Research Foundation, The BRCA Foundation,
actions, leading to the development of algorithms 20. H. E. Bryant et al., Nature 434, 913–917 (2005). and UCSF. We thank D. Krastev (ICR, London) for assistance with
that predict SL relationships (72). Integrating 21. A. Ashworth, C. J. Lord, J. S. Reis-Filho, Cell 145, 30–38 figure generation and A. Tutt (ICR, London and King’s College
functional genomics and proteomics with com- (2011). London) for helpful suggestions. A.A. is on the Scientific Advisory
putational network analysis approaches might 22. J. Murai et al., Cancer Res. 72, 5588–5599 (2012). Board of Genentech and is a paid consultant for AtlasMDX,
23. J. Murai et al., Mol. Cancer Ther. 13, 433–443 (2014). Third Rock Ventures, Pfizer, and Merck Serono. A.A. is a
therefore be useful to dissect these principles with 24. Y. Pommier, M. J. O’Connor, J. de Bono, Sci. Transl. Med. 8, cofounder of Tango Therapeutics. C.J.L. is a paid consultant for
the aim of streamlining the process of identifying 362ps17 (2016). AstraZeneca, Tango Therapeutics, and Sun Pharma. In accord with
highly penetrant SL effects with potential ther- 25. S. J. Pettitt et al., PLOS ONE 8, e61520 (2013). the Institute of Cancer Research Rewards to Inventors Scheme,
apeutic value. 26. J. E. Jaspers et al., Cancer Discov. 3, 68–81 (2013). A.A. is named as an inventor on patents describing the use of PARP
27. G. Xu et al., Nature 521, 541–544 (2015). inhibitors for the treatment of cancer [WO2014013231 (A1) no.
It took more than 10 years from the discovery 28. A. Ray Chaudhuri et al., Nature 535, 382–387 (2016). 2014-01-23, US2012135983 (A1) no. 2012-05-31, US2012010204 (A1)
of the PARPi/BRCA SL interaction to regulatory 29. S. L. Edwards et al., Nature 451, 1111–1115 (2008). no. 2012-01-12, US2006142231 (A1) no. 2006-06-29, and WO2008020180
approval. Now, based on preclinical studies re- 30. L. J. Barber et al., J. Pathol. 229, 422–429 (2013). (A2) no. 2008-02-21]. In accord with the Institute of Cancer
cently published and currently under way, PARPi 31. W. Sakai et al., Nature 451, 1116–1120 (2008). Research Rewards to Inventors Scheme, C.J.L. is named as an
remain a very active area of investigation. With 32. B. Norquist et al., J. Clin. Oncol. 29, 3008–3015 inventor on patents describing the use of PARP inhibitors for the
(2011). treatment of cancer [WO2014013231 (A1) no. 2014-01-23 and
the number of ongoing clinical trials, there is 33. C. J. Lord, A. Ashworth, Nat. Med. 19, 1381–1388 (2013). WO2008020180 (A2) no. 2008-02-21].
optimism that in the short term there will be 34. C. J. Lord, A. Ashworth, Nat. Rev. Cancer 16, 110–120
additional regulatory approvals for PARPi in mul- (2016). 10.1126/science.aam7344

Lord et al., Science 355, 1152–1158 (2017) 17 March 2017 6 of 6


PARP inhibitors: Synthetic lethality in the clinic
Christopher J. Lord and Alan Ashworth

Science 355 (6330), 1152-1158.


DOI: 10.1126/science.aam7344

Downloaded from http://science.sciencemag.org/ on December 25, 2020


ARTICLE TOOLS http://science.sciencemag.org/content/355/6330/1152

RELATED http://science.sciencemag.org/content/sci/355/6330/1142.full
CONTENT
http://science.sciencemag.org/content/sci/355/6330/1144.full
http://science.sciencemag.org/content/sci/355/6330/1147.full
http://science.sciencemag.org/content/sci/355/6330/1158.full
http://science.sciencemag.org/content/sci/355/6330/1163.full
http://science.sciencemag.org/content/sci/355/6330/1103.full
http://science.sciencemag.org/content/sci/355/6330/1131.full
http://stm.sciencemag.org/content/scitransmed/9/375/eaal2463.full
http://stm.sciencemag.org/content/scitransmed/8/362/362ps17.full
http://stke.sciencemag.org/content/sigtrans/7/326/ra47.full
http://stke.sciencemag.org/content/sigtrans/4/166/ra19.full
http://stke.sciencemag.org/content/sigtrans/10/480/eaam7479.full
REFERENCES This article cites 72 articles, 24 of which you can access for free
http://science.sciencemag.org/content/355/6330/1152#BIBL

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Use of this article is subject to the Terms of Service

Science (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement of
Science, 1200 New York Avenue NW, Washington, DC 20005. The title Science is a registered trademark of AAAS.
Copyright © 2017, American Association for the Advancement of Science

You might also like