You are on page 1of 10

Cancer

Review Research

Exploiting DNA Replication Stress for Cancer


Treatment
Tajinder Ubhi1,2 and Grant W. Brown1,2

Abstract
Complete and accurate DNA replication is fundamental to associated with such therapies. We discuss how replication
cellular proliferation and genome stability. Obstacles that stress modulates the cell-intrinsic innate immune response
delay, prevent, or terminate DNA replication cause the phe- and highlight the integration of replication stress with immu-
nomena termed DNA replication stress. Cancer cells exhibit notherapies. Together, exploiting replication stress for cancer
chronic replication stress due to the loss of proteins that treatment seems to be a promising strategy as it provides a
protect or repair stressed replication forks and due to the selective means of eliminating tumors, and with continuous

Downloaded from http://aacrjournals.org/cancerres/article-pdf/79/8/1730/2791709/1730.pdf by guest on 05 March 2023


continuous proliferative signaling, providing an exploitable advances in our knowledge of the replication stress response
therapeutic vulnerability in tumors. Here, we outline current and lessons learned from current therapies in use, we are
and pending therapeutic approaches leveraging tumor-specific moving toward honing the potential of targeting replication
replication stress as a target, in addition to the challenges stress in the clinic.

Introduction mental. In this review, we provide a summary of the therapies


centered on enhancing both endogenous and drug-induced rep-
The DNA replication machinery successfully carries out accu-
lication stress and discuss the rationales associated with them. We
rate genome duplication in the face of numerous obstacles, many
also highlight the potential of using replication stress to stimulate
of which cause DNA replication stress. Replication stress, defined
the cell-intrinsic innate immune response to improve immuno-
as any hindrance to DNA replication that either stalls, blocks, or
therapy effectiveness.
terminates DNA polymerization, activates a highly conserved
replication stress response pathway mediated by a network of
repair proteins to resolve the damage. Efficient removal of obsta- DNA Replication Stress: Causes,
cles to replication progression safeguards genome integrity by Consequences, and the Cellular Response
ensuring timely completion of DNA replication and by limiting
Eukaryotic DNA replication is a tightly regulated and complex
susceptibility to mutagenesis. However, failure to remove repli-
process that requires the orchestrated functions of several hun-
cation stressors due to the loss of replication stress response and
dred proteins during the S phase of each cell cycle (reviewed in
repair proteins and sustained proliferative signaling leads to
refs. 3, 4). Each time a cell divides, billions of nucleotides of DNA
chronic replication stress, and is a prominent feature of tumor
must be accurately polymerized, and ensuring this process occurs
cells. In particular, whole-genome sequencing efforts of tumor
prior to the next cell cycle is essential for cellular homeostasis.
samples have indicated the important role of replication stress
However, there are numerous hindrances to DNA replication that
in tumor progression and maintenance, complementing early
can prevent progression of the replication machinery and cause
findings from the groups of Halazonetis and Bartek who
replication forks to stall. Obstacles to replication fork progression
demonstrated that replication stress accumulates upon cellular
can arise from several sources, either endogenous or exogenous,
transformation, and is rarely observed in even the most prolifer-
ranging from the depletion of nucleotide pools available for
ative tissues (1, 2). Thus, due to the selective nature of replication
DNA synthesis to transcription–replication machinery collisions,
stress as a therapeutic target, the dependency of tumor cells on
RNA–DNA hybrids, oncogene-induced increases in replication
the replication stress response for cell survival, and the ever-
origin firing, and inherently hard-to-replicate regions (Fig. 1A;
expanding network of replication stress response proteins that
reviewed in ref. 5). The type of stress challenging replication
can be targeted, replication stress is beginning to be explored as an
fork progression dictates the cellular response and network of
attractive target in the clinic. In contrast to normal cells, cancer
repair proteins activated in response to the replication stress.
cells can be pushed toward cell death by introducing further DNA
For example, ribonucleotides that are misincorporated into
damage in a catastrophic manner or by promoting their entrance
replicating DNA can cause replication stress, and are recognized
into cell cycle stages where unresolved replication stress is detri-
and removed by ribonucleotide excision repair, whereas inter-
1
Department of Biochemistry, University of Toronto, 1 King's College Circle, strand crosslinks are mainly repaired by the Fanconi anemia
Toronto, Ontario, Canada. 2Donnelly Centre for Cellular and Biomolecular pathway, a specialized branch of the DNA damage response.
Research, University of Toronto, Toronto, Ontario, Canada.
Stalled replication forks are often transient, with cells able to
Corresponding Author: Grant W. Brown, University of Toronto, 160 College resolve them and continue replication. However, when left unre-
Street, Toronto, ON M5S 3E1, Canada. Phone: 416-946-5733; Fax: 416-978-8548; solved, replication stress can lead to small-scale and large-scale
E-mail: grant.brown@utoronto.ca
genome alterations such as the mutations and chromosomal
doi: 10.1158/0008-5472.CAN-18-3631 rearrangements found frequently in human cancers and devel-
2019 American Association for Cancer Research. opmental disorders.

1730 Cancer Res; 79(8) April 15, 2019


Exploiting Replication Stress

Replication stress triggers the activation of a signaling cascade surprising that replication stress is beginning to be rationally
that promotes resolution of the stress and resumption of repli- leveraged in cancer therapies. The elevated replication stress in
cation fork progression, collectively known as the S phase check- cancer cells has been largely attributed to the loss of cell cycle
point (Fig. 1B). Stalled replication forks are characterized by checkpoint activators, often tumor suppressor genes, and to the
stretches of single-stranded DNA (ssDNA), larger than those overexpression or constitutive expression of oncogenes (21, 23).
found during lagging strand DNA synthesis, that activate the These alterations modify replication fork rates, increase replica-
S phase checkpoint. It is thought that the ssDNA gaps are caused tion initiation or origin firing, and promote premature entry into
mainly by polymerase and helicase uncoupling (6), but ssDNA S phase (2, 13, 24). Strategies aiming to exploit replication stress
can also be generated by nucleolytic processing of DNA at reversed fall within two major categories; they either function to increase
replication fork structures (7). The ssDNA is bound with high the endogenous replication stress present within tumor cells to
affinity by replication protein A (RPA), which serves as a platform induce cell death or they produce tumor cell-specific replication
for the recruitment of numerous sensor proteins, including stress that can be further enhanced by additional therapeutics.
ataxia telangiectasia and Rad3-related (ATR)-interacting protein Traditional replication stress–inducing drugs largely resulted
(ATRIP), the 9-1-1 DNA clamp complex (RAD9-RAD1-HUS1), from serendipitous observations of tumor cell death following
topoisomerase II binding protein 1 (TOPBP1), and Ewing tumor- use of the agents (25), but increasingly drugs are designed with the
associated antigen 1 (ETAA1), which recruit and activate the specific goal of exploiting replication stress (Table 1). The increase
central replication stress response kinase ATR (reviewed in 8). in accessibility of deep sequencing approaches has provided

Downloaded from http://aacrjournals.org/cancerres/article-pdf/79/8/1730/2791709/1730.pdf by guest on 05 March 2023


Upon activation, ATR orchestrates a multifaceted response at remarkable insight into the catalogue of DNA repair genes mutat-
stalled replication forks by phosphorylating several downstream ed in cancers and offers another exciting means of targeting
targets, including its main effector kinase, checkpoint kinase 1 endogenous replication stress by identifying mutation-specific
(CHK1). The ATR–CHK1 signaling pathway leads to cell cycle vulnerabilities (26).
arrest, stabilization of stalled replication forks, and the inhibition
of late origin firing to prevent excess ssDNA formation (9). In Current Therapies That Harness
doing so, the S phase checkpoint ultimately promotes replication
fork repair and restart, or resumption of replication from an Replication Stress
adjacent origin (10, 11), to ensure complete replication of the Because a key feature that distinguishes tumor cells from
affected genomic loci. most normal cells is their sustained proliferation, DNA repli-
In instances where cells endure chronic replication stress, such cation has been harnessed as a semi-selective target in cancer
as in the early stages of tumorigenesis, or during extended periods therapies for decades (25). Many traditional anticancer drugs
of antineoplastic drug treatment, replication forks can col- act by increasing the replication stress within tumor cells, by
lapse (12–14). It is thought that ssDNA stretches found at stressed directly damaging the DNA, depleting cellular resources
replication forks are converted to DNA double-strand breaks required for successful cell division, or through a combination
during replication fork collapse and that the replication fork loses of both. Nucleoside analogues, including 5-fluorouracil, cytar-
its capacity to synthesize DNA. Although collapsed replication abine, and gemcitabine, were among the first chemotherapeutic
forks can likely be rescued through homologous recombina- agents introduced to the clinic and act through diverse mechan-
tion (15) or mitotic DNA synthesis (16), inability to resume isms to ultimately halt DNA replication. For instance, cytar-
DNA replication in these regions promotes mutagenesis and abine and gemcitabine are deoxycytidine analogues that com-
genome instability. Cells recruit error-prone translesion DNA pete directly with dCTP for incorporation into newly synthe-
polymerases that inaccurately synthesize DNA as a means to sized DNA, leading to a delay in replication fork progression or
complete bulk genome replication, failure of which leads to even a complete termination of some replication forks (27, 28).
incompletely replicated DNA persisting into mitosis. The presence In addition to their abilities to incorporate into DNA, these
of under-replicated genomic loci in mitosis can result in chro- agents also reduce the size of the intracellular nucleotide pools
mosomal entanglements between sister chromatids (17) or lag- available for DNA synthesis or limit the availability of a single
ging chromosomes that fail to segregate with the remaining nucleotide. For example, gemcitabine potentiates its cytotoxic
genetic material, leading to the formation of micronuclei (18). effects by also inhibiting ribonucleotide reductase (29), where-
These events not only increase the risk of genome instability as 5-fluorouracil functions mainly by inhibiting thymidylate
and promote transformation, but they also perpetuate the synthetase, thereby reducing the amount of thymidine avail-
existing genome instability through global missegregation of able for DNA replication and repair (30).
chromosomes, thus providing a route of evolution for cancer In contrast to nucleoside analogues, alkylating agents and
cells (19). Unresolved replication stress following mitosis leads platinum-based compounds function by directly modifying DNA
to the formation of nuclear bodies marked by the DNA damage through the formation of alkylation adducts on DNA bases, intra-
response protein p53 binding protein 1 (53BP1) in the subse- or interstrand crosslinks between DNA bases, or both (31, 32).
quent G1 phase of the cell cycle in daughter cells, which may act The generation of DNA adducts by alkylating agents such as
to protect or promote resolution of the replication stress (20). dacarbazine and temozolomide poses an obstacle for the
advancing DNA polymerase, leading to delayed replication fork
progression. Moreover, intra- and interstrand crosslinks generated
Exploiting DNA Replication Stress–Centered
by platinum-based agents such as cisplatin and carboplatin are
Vulnerabilities in Cancer physical barriers to DNA replication and block the replication
As it is well documented that tumor cells have persistent machinery. Intrastrand crosslinks present on the template DNA
replication stress (21), and with replication stress now acknowl- strand during replication impair proper base pairing with the
edged as an enabling characteristic of cancer (22), it is not nascent DNA strand, whereas interstrand crosslinks impede

www.aacrjournals.org Cancer Res; 79(8) April 15, 2019 1731


Ubhi and Brown

A RNA processing enzymes,


Regulation of RNA-DNA helicases,
replication origin and topoisomerases
firing
Oncogene-induced
replication stress
RNA-DNA hybrids
Nucleotide pool MYC and replication- DNA sliding clamp
depletion transcription conflicts DNA polymerase
C DNA helicase
A H3N Cl
T G
Pt
H3N Cl

Inter-strand
GCGG crosslinks

OH Hard-to-replicate
regions Fanconi Anemia
Ribonucleotide pathway
incorporation

Downloaded from http://aacrjournals.org/cancerres/article-pdf/79/8/1730/2791709/1730.pdf by guest on 05 March 2023


DNA helicases
Ribonucleotide
excision repair

9-1-1 DNA clamp complex


RPA DNA lesion

ATRIP ATRIP
TOPBP1 ATR ATR ETAA1
P

CHK1

Stabilization of Late origin firing


stalled replication Cell cycle
forks progression
C
AZD1775 WEE1 G1: Gap phase I
S: Synthesis phase
G2: Gap phase II
M: Cell division phase
G2/M checkpoint

M
G1
G2
S
p53

Prexasertib,
GDC-0575,
SCH 900776, G1/S checkpoint
and SRA737

S phase
CHK1 ATR checkpoint

M6620, AZD6738,
and BAY1895344
© 2019 American Association for Cancer Research

1732 Cancer Res; 79(8) April 15, 2019 Cancer Research


Exploiting Replication Stress

the very first step of DNA replication, that is, unwinding of the target cell cycle checkpoints (Fig. 1C) to ultimately promote the
double-strand helix (33). Another class of replication stress– premature entry of tumor cells into mitosis, where they undergo
inducing agents that directly damage DNA is topoisomerase cell death from abnormal mitosis, termed mitotic catastro-
inhibitors (34, 35). Topoisomerases generate transient DNA phe (53). Most importantly, these approaches promise an
breaks to relax DNA supercoiling that occurs during DNA improved and practical therapeutic index relative to conventional
replication and transcription. The intermediate state where the agents due to the intrinsically high levels of replication stress often
topoisomerases are bound to the cleaved DNA is trapped by found within tumor cells.
topoisomerase inhibitors and leads to the formation of stable Activation of the S phase checkpoint elicits a multifaceted
protein–DNA complexes that generate a physical barrier to ongo- signaling response, where each pathway can provide additional
ing replication forks, often collapsing to double-strand breaks targets for therapeutic intervention (Fig. 1). The S phase check-
when encountered by a replication fork. The action of topoisom- point response kinases ATR and CHK1 have been the leading
erase inhibitors resembles that of another group of replication targets of the therapies currently under investigation (Table 1).
stress–inducing therapeutics, PARP inhibitors. PARP inhibitors ATR delays the activation of dormant replication origins and
generate an obstacle for the replication machinery by trapping stabilizes stressed replication forks to prevent replication fork
PARP on DNA (36), in addition to impairing single-strand break collapse. Accordingly, ATR inhibition initiates widespread DNA
repair (37) and accelerating replication fork progression (38), synthesis from normally dormant replication origins, generat-
amplifying the replication stress in tumor cells. ing enough ssDNA to exhaust the cellular pools of RPA and

Downloaded from http://aacrjournals.org/cancerres/article-pdf/79/8/1730/2791709/1730.pdf by guest on 05 March 2023


The replication stress–inducing effects of conventional thera- depleting the nucleotide pools necessary for DNA synthesis (9,
pies have been directly visualized at the single-molecule level 11). ATR and CHK1 inhibition also promotes premature entry
through DNA fiber analyses, where DNA synthesis rates of indi- into mitosis despite the presence of under-replicated DNA,
vidual DNA replication forks can be measured following drug leading to chromosome fragmentation (54, 55). Given their
treatment (39–41), and through DNA pull-down techniques mechanisms of action, ATR and CHK1 inhibitors synergize
that identify proteins bound directly at replication forks with compounds that induce replication stress, and increase
in vivo (42–46). Of particular interest, the interactions between the toxicity of nucleoside analogues in ovarian, lung, and pan-
traditional replication stress–inducing drugs and cellular replica- creatic cancer (56–60), platinum-based agents in lung, gastric,
tion stress response pathways are evident in preclinical studies, ovarian, bladder, and breast cancer (50, 61, 62) and topoisom-
where inhibition or depletion of the key replication stress erase inhibitors in breast, colon, and brain tumors (63, 64).
response kinases required for overcoming the replication stress Because ATR is activated following replication stress to ensure
present within cells, ATR and/or CHK1, sensitizes tumor cells to successful resolution of the replication stress, ATR inhibitors
5-fluorouracil, gemcitabine, cisplatin, PARP inhibitors, and temo- should be sufficient as monotherapy in tumors that experience
zolomide, with some combinations having advanced to clinical high levels of intrinsic replication stress, which is also a current
trials (34, 35, 47–52). arm of investigation (NCT03188965). Early CHK1 inhibitors
were not successful as monotherapies, largely due to their intol-
Emerging Combination Therapies That erable side effects and pharmacokinetic properties, although
clinical studies demonstrate potential for newer generation
Target Replication Stress: Cell Cycle CHK1 inhibitors, as they have a tolerable toxicity profile (65).
Checkpoints The high toxicity of CHK1 inhibitors is consistent with the central
Although conventional replication stress–inducing agents have role of CHK1 in mediating the S phase checkpoint upon replica-
been used for decades, their applicability and efficacy is limited by tion stress, both in the absence and presence of ATR, and
their associated toxicities and the rapid emergence of resistant in preventing premature entry into mitosis (55, 66), but presents
tumor cells. As noted, these therapies target DNA replication, and the possibility of targeting downstream effectors of CHK1
thus have an effect on highly proliferative cells, including normal therapeutically.
cells within the gut epithelium and bone marrow, leading to As tumor cells are often deficient in the key G1/S transition
undesirable and sometimes intolerable side effects. In light of this, regulator p53, the G2/M checkpoint becomes essential to prevent
several promising rationally designed combination therapies that tumor cells with unreplicated or damaged DNA from entering
increase the specificity of these traditional therapies toward tumor catastrophic mitoses. Promising therapies targeting regulators of
cells are currently undergoing clinical trials. In addition to gen- mitotic entry to undermine the G2/M checkpoint are also being
erating further replication stress, many of these therapies also explored. Inhibitors of the G2/M checkpoint kinase WEE1 have

Figure 1.
Generation and exploitation of DNA replication stress. A, Obstacles in replication fork progression can arise from exogenous and endogenous sources and
dictate the cellular response and network of repair proteins activated in response to the replication stress. Sources of replication stress include, but are not
limited to, the depletion of nucleotide pools available for DNA replication and repair, oncogene-induced stress, RNA–DNA hybrids, replication–transcription
conflicts, DNA crosslinks, inherently difficult-to-replicate regions, and the misincorporation of ribonucleotides into replicating DNA. B, Replication stress
activates a highly conserved signaling response called the S phase checkpoint. The S phase checkpoint is activated upon exposure of ssDNA from stressed
replication forks, which leads to the recruitment and activation of the central replication stress response kinase ATR and its main effector kinase, CHK1. The ATR–
CHK1 signaling pathway promotes cell cycle arrest, stabilization of stalled replication forks, and inhibition of late origin firing to ultimately ensure complete
replication of the affected genomic loci. C, Cancer cells become dependent on the S phase checkpoint for cell survival due to their high levels of intrinsic DNA
replication stress. In addition to S phase checkpoint response proteins being the leading targets for cancer therapies targeting replication stress, regulators of
mitotic entry are also being evaluated, as they drive cells with unresolved replication stress into catastrophic mitoses. Targets in the DNA replication stress
response are shown for each cell cycle checkpoint, with inhibitors targeting those proteins currently being evaluated in clinical trials in bold.

www.aacrjournals.org Cancer Res; 79(8) April 15, 2019 1733


Ubhi and Brown

Table 1. Chemotherapeutics that increase DNA replication stress


Class of agents or target Function Compounds Clinical stage References
Nucleoside analogues Inhibition of DNA replication Azacitidine All approved for use 30, 109
Cytarabine
Decitabine
Gemcitabine
5-Fluorouracil
Alkylating agents and Direct modification of DNA Carboplatin All approved for use 31, 32
platinum compounds Cisplatin
Cyclophosphamide
Dacarbazine
Methotrexate
Mitomycin C
Oxaliplatin
Procarbazine
Temozolomide
Topoisomerase I and II Relax DNA supercoiling that occurs during Belotecan Approved 34, 35
DNA replication and transcription CRLX101 Phase II
Doxorubicin Approved
Epirubicin Approved

Downloaded from http://aacrjournals.org/cancerres/article-pdf/79/8/1730/2791709/1730.pdf by guest on 05 March 2023


Etoposide Approved
Idarubicin Approved
Irinotecan Approved
LMP400 Phase I
LMP776 Phase I
Mitoxantrone Approved
NKTR-102 Phase III
Teniposide Approved
Topotecan Approved
PARP Single-strand DNA break repair Olaparib Approved 110
Niraparib Approved
Rucaparib Approved
Talazoparib Approved
Veliparib Phase III
ATR Central replication stress response kinase AZD6738 Phase I/II 52
BAY1895344 Phase I/II
M6620 Phase II
CHK1 Main effector kinase of ATR in replication GDC-0575 Phase I 111
stress response SCH 900776 Phase I
SRA737 Phase I
Prexasertib Phase I/II
WEE1 G2/M checkpoint kinase AZD1775 Phase I/II 112

shown promising results with an acceptable toxicity profile (67). Precision Medicine Approaches That
Drugs that increase replication stress should increase the impor-
Leverage Replication Stress
tance of the G2/M checkpoint for tumor cell survival, and accord-
ingly there are currently greater than 30 clinical studies underway The identification of the underlying genetic alterations across
for combination therapies with WEE1 inhibitors (clinicaltrials. all major subtypes of cancer has provided unprecedented insight
gov, accessed January 2019). These include combinations of into DNA repair proteins that are altered in cancer (26) and has
gemcitabine, temozolomide, cytarabine, irinotecan, carboplatin, laid the foundation for several precision medicine approaches.
and cisplatin with the potent WEE1 inhibitor AZD1775 in Almost every tumor has at least one gene in the replication stress
pancreatic, blood, brain, ovarian, and colon tumors, with an response altered, providing ample opportunities to exploit tumor-
emphasis on p53 deficiency as the key selection criteria. In specific alterations without affecting normal cells. The concept of
addition to driving tumor cells into unscheduled mitosis (68), synthetic lethality (73), whereby the presence of cancer-specific
the cytotoxicity of AZD1775 could also be attributed to the mutations in specific genes renders other genes essential for cell
effects WEE1 has on stabilizing stalled replication forks and proliferation and survival, has been fundamental for the discovery
ensuring timely origin firing, further exacerbating the replica- of several of these gene-targeted strategies (74). Perhaps the best
tion stress in tumor cells when WEE1 is inhibited (69, 70). example of this strategy is the successful use of PARP inhibitors for
WEE1 inhibitors act synergistically with ATR and CHK1 inhi- the treatment of BRCA1/BRCA2-deficient breast and ovarian
bitors in preclinical studies (71, 72), extending the catalogue of cancers, as these double-strand break repair defective tumors are
possible combinations with other targeted inhibitors that dependent on PARP activity for cell survival (37, 75). Consider-
could be implemented. Similar to ATR and CHK1 inhibitors, able effort is now being expended to identify additional synthetic
although AZD1775 has shown single-agent activity in preclin- lethal interactions with DNA repair genes, including replication
ical studies, its efficacy is potentiated when integrated into stress response genes. One of the most promising targets
combination regimens. currently being explored is ATR. Tumors harboring mutations in

1734 Cancer Res; 79(8) April 15, 2019 Cancer Research


Exploiting Replication Stress

genes encoding the chromatin remodeler ARID1A or the DNA or WEE1 activity is lethal in the context of cyclin E-, MYC-, and/or
damage kinase ATM are synthetically sensitive to ATR inhibitors, RAS-overexpressing tumor cells (84–87), presumably because
with a complete response observed in a patient with metastatic these tumors have a heightened requirement for the cell cycle
ATM-deficient colorectal cancer (76–78). The dependency of checkpoints that respond to replication stress. Overexpression
ARID1A-deficient tumors on ATR has been attributed to the of the G1/S checkpoint regulator, cyclin D and its effectors
mitotic defects present in these cells, which causes an increased CDK4 and CDK6, are leveraged as biomarkers for patient selection
reliance on ATR function. Both ARID1A and ATM are frequently in trials with DNA damaging agents and immunotherapies
altered in tumors, particularly in ovarian and endometrial cancers (NCT03237390, NCT02290145, and NCT03088059). More
for ARID1A and lymphoid malignancies for ATM, illustrating the recently, studies have also elucidated the role of apolipoprotein
applicability of these findings. In addition to further interactions B mRNA editing enzyme catalytic polypeptide–like (APOBEC)
with ATR, tumors containing mutations in other frequently proteins as an endogenous source of replication stress and have
altered replication stress response genes including RPA1 found that overexpression of two APOBEC3 enzymes, APOBEC3A
(NCT03207347 and NCT03377556) and the mutagenic DNA and APOBEC3B, confers sensitivity to ATR inhibition (88, 89). As
polymerase POLQ (79) are currently being explored to identify APOBEC3A and APOBEC3B are overexpressed in several cancer
druggable synthetic lethal interactions. Genetic interactions types (90, 91), this finding could have important implications in
with replication stress–inducing agents including CHK1 (80, maximizing the potential of ATR inhibitors while sparing normal
81) and WEE1 (82) inhibitors are also being investigated for cells. The combination of the overexpression of the remaining five

Downloaded from http://aacrjournals.org/cancerres/article-pdf/79/8/1730/2791709/1730.pdf by guest on 05 March 2023


use in mutation-specific tumor backgrounds, such as the use of APOBEC3 paralogs and replication stress–inducing agents may,
CHK1 inhibitors in EZH2-deficient T-cell acute lymphoblastic therefore, also be worth testing for possible synergies.
leukemias.
Patient-tailored therapies targeting vulnerabilities induced by The Innate Immune Response: An Emerging
specific oncogenes are also being implemented. In particular,
the genes encoding RAS, cyclin E, cyclin D, and MYC are frequently Marker of the Replication Stress Response
amplified in many cancer types and are starting to be harnessed An exciting emerging concept is the link between replication
as biomarkers of replication stress. These oncogenes stimulate stress and activation of the cell-intrinsic innate immune response
premature entry into S phase, thereby disrupting the spatial (Fig. 2). Accumulating evidence indicates that replication stress–
and temporal regulation of origin firing and replication fork inducing agents such as topoisomerase inhibitors and cells defi-
speed, increase global transcription activity, and promote cient in replication stress response genes induce the expression
RNA–DNA hybrid accumulation, ultimately causing replication of type I IFNs and proinflammatory cytokines (92–95). It is now
stress (reviewed in ref. 83). Accordingly, inhibition of ATR, CHK1, apparent that this activation is largely mediated through the

Cytosol
Replication stress conditions
TREX1
RPA and
RAD51 Type I inteferon production
cGAS and
IFI16 STING
Interferon-stimulated gene
and pro-inflammatory cytokine
SAMHD1 production

STING Cellular senescence or


Cytosolic DNA activation elimination by adaptive
sensing immune system

Nucleus

Nuclear envelope
Micronucleus

© 2019 American Association for Cancer Research

Figure 2.
Activation of the cell-intrinsic innate immune response by DNA replication stress. In replication stress conditions, nuclear DNA is released into the cytosol, either
directly from stalled replication forks in three prime repair exonuclease 1 (TREX1)-, sterile alpha motif and HD domain–containing protein 1 (SAMHD1)-, RPA- or
RAD51-deficient backgrounds, or from micronuclei, which are small nuclei formed predominantly following mitotic progression in cells experiencing genome
instability. The cytosolic DNA fragments are sensed by the pattern recognition receptors, cyclic GMP-AMP synthase (cGAS) and IFNg-inducible factor 16 (IFI16),
which activate the central adaptor protein STING, inducing a transcriptional response to ultimately promote cellular senescence or elimination by the adaptive
immune system.

www.aacrjournals.org Cancer Res; 79(8) April 15, 2019 1735


Ubhi and Brown

presence of cytosolic DNA derived from the nucleus or mitochon- replication stress. Knowledge of the replication stress response
dria, which engages the pattern recognition receptors that nor- continues to develop as we discover novel proteins and reveal
mally scan for viral infections. The mechanisms through which intricate response networks, providing greater opportunities to
replication stress induces DNA release into the cytoplasm appear target these proteins in therapy. In particular, advances in molec-
to be specific to the perturbation. Recent work has found micro- ular biology tools continue to facilitate identification of novel
nuclei to be a key source of immunostimulatory DNA following molecular targets in the replication stress response and the inter-
mitotic progression in cells lacking RNase H2 (96, 97), whereas play between the replication stress response and different cellular
the release of ssDNA directly from stalled replication forks has pathways such as metabolism (104). In addition to RNA inter-
also been demonstrated in TREX1-, SAMHD1-, RPA-, and RAD51- ference screens, the advent of genome-wide screening using the
deficient backgrounds and following cytarabine treatment CRISPR-Cas9 technology will provide a powerful tool moving
(93, 98–101). Whether these conditions could result in cyto- forward to rapidly identify novel clinically relevant synthetic
plasmic DNA accumulation through both observed mechanisms lethal interactions in cancer cells, and is already yielding prom-
has yet to be ruled out, and several aspects of these mechanisms, ising results (105–108). As these technologies begin to be adapted
including the interplay of proteins at replication forks that pro- in other systems, similar screens performed in more biologically
mote ssDNA release, have yet to be defined. appropriate settings, such as in 3D organoid cultures or in vivo
Central to the innate immune response is the adaptor protein mouse models, will likely yield more clinically relevant synthetic
"stimulator of IFN genes" (STING), which couples signals from lethal interactions. Furthermore, advances in whole-genome

Downloaded from http://aacrjournals.org/cancerres/article-pdf/79/8/1730/2791709/1730.pdf by guest on 05 March 2023


cytosolic DNA sensors to a transcriptional response from activa- sequencing of tumors will also assist precision medicine
tion of both the NF-kB and type I IFN signaling axes, ultimately approaches by identifying key actionable genetic alterations for
promoting cellular senescence or elimination by the adaptive therapeutic targeting in the clinic.
immune system (reviewed in ref. 102). Interestingly, STING Despite the unprecedented rate at which we are identifying
signaling is suppressed in several tumors (103) and multiple novel replication stress response targets, there remain several
cancer cell types contain genome-derived cytosolic ssDNA (100), obstacles to overcome for optimal treatment outcomes. One
further affirming the presence and importance of persistent rep- limitation of all replication stress–targeting therapies that will
lication stress in tumors. As type I IFN production from the innate be difficult to diminish is achieving an acceptable therapeutic
response is critical in priming the adaptive immune system, window, due to the essentiality of the replication stress
robust STING signaling has been attributed to an increased response in normal cells. In contrast to conventional therapies
immunotherapy response. In fact, blocking T-cell inhibitory path- that target all dividing cells, targeted inhibitors have shown
ways using immune checkpoint inhibitors is ineffective in mice greater precision in selectively affecting tumors, yet they con-
lacking TMEM173 (STING; ref. 96). However, despite current tinue to present adverse side effects that are sometimes intol-
efforts to combine STING agonists with checkpoint inhibitors as erable. Another important limitation is the current technolo-
antitumor therapy, therapeutic delivery of the agonists has yet to gies available to predict tumor backgrounds that would benefit
be improved for clinical settings. Combination approaches inte- most from certain therapies. Markers that can be effectively
grating replication stress–inducing agents, such as carboplatin detected by immunohistochemistry, such as the overexpres-
or gemcitabine, with immunotherapies like the immune check- sion of certain oncogenes, are starting to be implemented as a
point inhibitor nivolumab, have advanced to clinical trials means to stratify patients for targeted treatment strategies in
(NCT02944396, NCT03662074, NCT03061188, NCT02734004, clinical trials. However, the dependence on immunohistolo-
NCT02849496, NCT02657889, and NCT02571725). It is tempt- gical approaches for biomarker identification restricts the use
ing to speculate that the efficacy observed could be due to the of important replication stress markers such as RPA foci and
engagement of STING by replication stress, and would support ssDNA and the use of functional DNA repair assays as proxies
the use of replication stress as a predictive marker for immu- for therapy response.
notherapy efficacy. Further work is required to better under- The notion of exploiting intrinsic replication stress has driven
stand the interplay between the host immune systems and considerable excitement in the medical oncology field as it pro-
replication stress and would provide insight into how these vides a selective means of eliminating tumors. Several approaches
responses can be modulated optimally. Interestingly, somatic undergoing clinical investigation are already showing promise,
mutations in the genes encoding the replicative polymerases and with the advent of powerful new technologies, both in
POLD1 and POLE are also being used as indicators for success- academic and clinical settings, many more are likely to be revealed
ful immunotherapy outcomes due to the high mutation for therapeutic development.
burden present in these genetic backgrounds (NCT03375307,
NCT03207347, NCT03428802, and NCT03491345). The role Disclosure of Potential Conflicts of Interest
of the innate response in this interaction has yet to be revealed No potential conflicts of interest were disclosed.
and will likely also provide additional exploitable therapeutic
vulnerabilities. Acknowledgments
Work in the authors' lab is supported by the Canadian Cancer Society, the
Canadian Institutes of Health Research, and an Ontario Graduate Scholarship
Future Directions (to T. Ubhi). We thank Haley Wyatt and Dan Durocher for helpful discussions
and careful reading of the manuscript.
Our knowledge of replication stress–centered vulnerabilities in
tumor cells has grown dramatically in recent years and is leading Received November 19, 2018; revised January 8, 2019; accepted January 28,
to an increasingly complex view of how tumor cells deal with 2019; published first April 9, 2019.

1736 Cancer Res; 79(8) April 15, 2019 Cancer Research


Exploiting Replication Stress

References
1. Gorgoulis VG, Vassiliou LV, Karakaidos P, Zacharatos P, Kotsinas A, 26. Knijnenburg TA, Wang L, Zimmermann MT, Chambwe N, Gao GF,
Liloglou T, et al. Activation of the DNA damage checkpoint and genomic Cherniack AD, et al. Genomic and molecular landscape of DNA damage
instability in human precancerous lesions. Nature 2005;434:907–13. repair deficiency across The Cancer Genome Atlas. Cell Rep 2018;23:
2. Bartkova J, Hor ejsí Z, Koed K, Kr€amer A, Tort F, Zleger K, et al. DNA 239–54.
damage response as a candidate anti-cancer barrier in early human 27. Richardson KA, Vega TP, Richardson FC, Moore CL, Rohloff JC, Tomkinson
tumorigenesis. Nature 2005;434:864–70. B, et al. Polymerization of the triphosphates of AraC, 20 ,20 -difluorodeox-
3. Fragkos M, Ganier O, Coulombe P, Mechali M. DNA replication origin ycytidine (dFdC) and OSI-7836 (T-araC) by human DNA polymerase a
activation in space and time. Nat Rev Mol Cell Biol 2015;16:360–74. and DNA primase. Biochem Pharmacol 2004;68:2337–46.
4. Burgers PMJ, Kunkel TA. Eukaryotic DNA replication fork. Annu Rev 28. Huang P, Chubb S, Hertel LW, Grindey GB, Plunkett W. Action of 20 ,20 -
Biochem 2017;86:417–38. difluorodeoxycytidine on DNA synthesis. Cancer Res 1991;51:6110–7.
5. Zeman M, Cimprich K. Causes and consequences of replication stress. 29. Cerqueira NM, Fernandes PA, Ramos MJ. Understanding ribonucleotide
Nat Cell Biol 2014;16:2–9. reductase inactivation by gemcitabine. Chemistry 2007;13:8507–15.
6. Byun TS, Pacek M, Yee MC, Walter JC, Cimprich KA. Functional uncou- 30. Longley DB, Harkin DP, Johnston PG. 5-fluorouracil: mechanisms of
pling of MCM helicase and DNA polymerase activities activates the action and clinical strategies. Nat Rev Cancer 2003;3:330–8.
ATR-dependent checkpoint. Genes Dev 2005;19:1040–52. 31. Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs.
7. Zellweger R, Dalcher D, Mutreja K, Berti M, Schmid JA, Herrador R, et al. Nat Rev Drug Discov 2005;4:307–20.
Rad51-mediated replication fork reversal is a global response to genotoxic 32. Fu D, Calvo JA, Samson LD. Balancing repair and tolerance of DNA
treatments in human cells. J Cell Biol 2015;208:563–79. damage caused by alkylating agents. Nat Rev Cancer 2012;12:104–20.
8. Saldivar JC, Cortez D, Cimprich KA. The essential kinase ATR: Ensuring 33. Deans AJ, West SC. DNA interstrand crosslink repair and cancer. Nat Rev

Downloaded from http://aacrjournals.org/cancerres/article-pdf/79/8/1730/2791709/1730.pdf by guest on 05 March 2023


faithful duplication of a challenging genome. Nat Rev Mol Cell Biol 2017; Cancer 2011;11:467–80.
18:622–36. 34. Pommier Y. Topoisomerase I inhibitors: Camptothecins and beyond.
9. Toledo LI, Altmeyer M, Rask MB, Lukas C, Larsen DH, Povlsen LK, et al. Nat Rev Cancer 2006;6:789–802.
ATR prohibits replication catastrophe by preventing global exhaustion of 35. Nitiss JL. Targeting DNA topoisomerase II in cancer chemotherapy.
RPA. Cell 2013;155:1088–103. Nat Rev Cancer 2009;9:338–50.
10. Ge XQ, Blow JJ. Chk1 inhibits replication factory activation but allows 36. Str€
om CE, Johansson F, Uhlen M, Szigyarto CAK, Erixon K, Helleday T.
dormant origin firing in existing factories. J Cell Biol 2010;191:1285–97. Poly (ADP-ribose) polymerase (PARP) is not involved in base excision
11. Chen YH, Jones MJK, Yin Y, Crist SB, Colnaghi L, Sims RJ, et al. ATR- repair but PARP inhibition traps a single-strand intermediate.
mediated phosphorylation of FANCI regulates dormant origin firing in Nucleic Acids Res 2011;39:3166–75.
response to replication stress. Mol Cell 2015;58:323–38. 37. Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, et al.
12. Petermann E, Orta ML, Issaeva N, Schultz N, Helleday T. Hydroxyurea- Targeting the DNA repair defect in BRCA mutant cells as a therapeutic
stalled replication forks become progressively inactivated and require two strategy. Nature 2005;434:917–21.
different RAD51-mediated pathways for restart and repair. Mol Cell 2010; 38. Maya-Mendoza A, Moudry P, Merchut-Maya JM, Lee M, Strauss R, Bartek J.
37:492–502. High speed of fork progression induces DNA replication stress and
13. Macheret M, Halazonetis TD. Intragenic origins due to short G1 phases genomic instability. Nature 2018;559:279–84.
underlie oncogene-induced DNA replication stress. Nature 2018;555: 39. Merrick CJ, Jackson D, Diffley JF. Visualization of altered replication
112–6. dynamics after DNA damage in human cells. J Biol Chem 2004;279:
14. Cortez D. Preventing replication fork collapse to maintain genome 20067–75.
integrity. DNA Repair 2015;32:149–57. 40. Kopper F, Bierwirth C, Schon M, Kunze M, Elvers I, Kranz D, et al. Damage-
15. Costantino L, Sotiriou SK, Rantala JK, Magin S, Mladenov E, Helleday T, induced DNA replication stalling relies on MAPK-activated protein kinase
et al. Break-induced replication repair of damaged forks induces genomic 2 activity. Proc Natl Acad Sci U S A 2013;110:16856–61.
duplications in human cells. Science 2014;343:88–91. 41. Hagenkort A, Paulin CBJ, Desroses M, Sarno A, Wiita E, Mortusewicz O,
16. Minocherhomji S, Ying S, Bjerregaard VA, Bursomanno S, Aleliunaite A, et al. dUTPase inhibition augments replication defects of 5-Fluorouracil.
Wu W, et al. Replication stress activates DNA repair synthesis in mitosis. Oncotarget 2017;8:23713–26.
Nature 2015;528:286–90. 42. Ribeyre C, Zellweger R, Chauvin M, Bec N, Larroque C, Lopes M, et al.
17. Gisselsson D, Pettersson L, Hoglund M, Heidenblad M, Gorunova L, Nascent DNA proteomics reveals a chromatin remodeler required for
Wiegant J, et al. Chromosomal breakage-fusion-bridge events cause topoisomerase I loading at replication forks. Cell Rep 2016;15:300–9.
genetic intratumor heterogeneity. Proc Natl Acad Sci U S A 2000;97: 43. Sirbu BM, Couch FB, Cortez D. Monitoring the spatiotemporal dynamics
5357–62. of proteins at replication forks and in assembled chromatin using isola-
18. Crasta K, Ganem NJ, Dagher R, Lantermann AB, Ivanova EV, Pan Y, et al. tion of proteins on nascent DNA. Nat Protoc 2012;7:594–605.
DNA breaks and chromosome pulverization from errors in mitosis. 44. Lossaint G, Larroque M, Ribeyre C, Bec N, Larroque C, Decaillet C, et al.
Nature 2012;482:53–8. FANCD2 binds MCM proteins and controls replisome function upon
19. Zhang CZ, Spektor A, Cornils H, Francis JM, Jackson EK, Liu S, et al. activation of S phase checkpoint signaling. Mol Cell 2013;51:678–90.
Chromothripsis from DNA damage in micronuclei. Nature 2015;522: 45. Dungrawala H, Rose KL, Bhat KP, Mohni KN, Glick GG, Couch FB, et al.
179–84. The replication checkpoint prevents two types of fork collapse without
20. Lukas C, Savic V, Bekker-Jensen S, Doil C, Neumann B, Pedersen RS, et al. regulating replisome stability. Mol Cell 2015;59:998–1010.
53BP1 nuclear bodies form around DNA lesions generated by mitotic 46. Alabert C, Bukowski-Wills JC, Lee SB, Kustatscher G, Nakamura K, De
transmission of chromosomes under replication stress. Nat Cell Biol Lima Alves F, et al. Nascent chromatin capture proteomics determines
2011;13:243–53. chromatin dynamics during DNA replication and identifies unknown
21. Macheret M, Halazonetis TD. DNA replication stress as a hallmark of fork components. Nat Cell Biol 2014;16:281–93.
cancer. Annu Rev Pathol 2015;10:425–48. 47. Fujinaka Y, Matsuoka K, Iimori M, Tuul M, Sakasai R, Yoshinaga K, et al.
22. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell ATR-Chk1 signaling pathway and homologous recombinational repair
2011;144:646–74. protect cells from 5-fluorouracil cytotoxicity. DNA Repair 2012;11:
23. Halazonetis TD, Gorgoulis VG, Bartek J. An oncogene-induced DNA 247–58.
damage model for cancer development. Science 2008;319:1352–5. 48. Calvo E, Braiteh F, Von Hoff D, McWilliams R, Becerra C, Galsky MD, et al.
24. Bester AC, Roniger M, Oren YS, Im MM, Sarni D, Chaoat M, et al. Phase I study of CHK1 inhibitor LY2603618 in combination with
Nucleotide deficiency promotes genomic instability in early stages of gemcitabine in patients with solid tumors. Oncology 2016;91:251–60.
cancer development. Cell 2011;145:435–46. 49. Kim H, George E, Ragland RL, Rafail S, Zhang R, Krepler C, et al. Targeting
25. Cheung-Ong K, Giaever G, Nislow C. DNA-damaging agents in cancer the ATR/CHK1 axis with PARP inhibition results in tumor regression
chemotherapy: serendipity and chemical biology. Chem Biol 2013;20: in BRCA-mutant ovarian cancer models. Clin Cancer Res 2017;23:
648–59. 3097–108.

www.aacrjournals.org Cancer Res; 79(8) April 15, 2019 1737


Ubhi and Brown

50. Mohni KN, Thompson PS, Luzwick JW, Glick GG, Pendleton CS, genome through control of replication initiation and nucleotide con-
Lehmann BD, et al. A synthetic lethal screen identifies DNA repair sumption. Mol Cell Biol 2012;32:4226–36.
pathways that sensitize cancer cells to combined ATR inhibition and 71. Jin J, Fang H, Yang F, Ji W, Guan N, Sun Z, et al. Combined inhibition of
cisplatin treatments. PLoS One 2015;10:e0125482. ATR and WEE1 as a novel therapeutic strategy in triple-negative breast
51. Eich M, Roos WP, Nikolova T, Kaina B. Contribution of ATM and ATR to cancer. Neoplasia 2018;20:478–88.
the resistance of glioblastoma and malignant melanoma cells to the 72. Hauge S, Naucke C, Hasvold G, Joel M, Rødland GE, Juzenas P, et al.
methylating anticancer drug temozolomide. Mol Cancer Ther 2013;12: Combined inhibition of Wee1 and Chk1 gives synergistic DNA damage in
2529–40. S-phase due to distinct regulation of CDK activity and CDC45 loading.
52. Lecona E, Fernandez-Capetillo O. Targeting ATR in cancer. Nat Rev Cancer Oncotarget 2017;8:10966–79.
2018;18:586–95. 73. Dobzhansky T. Genetics of natural populations; recombination and
53. Mansilla S, Bataller M, Portugal J. Mitotic catastrophe as a consequence of variability in populations of Drosophila pseudoobscura. Genetics
chemotherapy. Anticancer Agents Med Chem 2006;6:589–602. 1946;31:269–90.
54. Eykelenboom JK, Harte EC, Canavan L, Pastor-Peidro A, Calvo-Asensio I, 74. O'Neil NJ, Bailey ML, Hieter P. Synthetic lethality and cancer. Nat Rev
Llorens-Agost M, et al. ATR activates the S-M checkpoint during unper- Genet 2017;18:613–23.
turbed growth to ensure sufficient replication prior to mitotic onset. 75. Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, et al.
Cell Rep 2013;5:1095–107. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-
55. Suzuki M, Yamamori T, Bo T, Sakai Y, Inanami O. MK-8776, a novel Chk1 ribose) polymerase. Nature 2005;434:913–7.
inhibitor, exhibits an improved radiosensitizing effect compared to 76. Williamson CT, Miller R, Pemberton HN, Jones SE, Campbell J, Konde A,
UCN-01 by exacerbating radiation-induced aberrant mitosis. Transl Oncol et al. ATR inhibitors as a synthetic lethal therapy for tumours deficient in
2017;10:491–500. ARID1A. Nat Commun 2016;7:13837.

Downloaded from http://aacrjournals.org/cancerres/article-pdf/79/8/1730/2791709/1730.pdf by guest on 05 March 2023


56. Hall AB, Newsome D, Wang Y, Boucher DM, Eustace B, Gu Y, et al. 77. Brown JS, O'Carrigan B, Jackson SP, Yap TA. Targeting DNA repair in
Potentiation of tumor responses to DNA damaging therapy by the cancer: beyond PARP inhibitors. Cancer Discov 2017;7:20–37.
selective ATR inhibitor VX-970. Oncotarget 2014;5:5674–85. 78. Kwok M, Davies N, Agathanggelou A, Smith E, Oldreive C, Petermann E,
57. Liu S, Ge Y, Wang T, Edwards H, Ren Q, Jiang Y, et al. Inhibition of ATR et al. ATR inhibition induces synthetic lethality and overcomes chemore-
potentiates the cytotoxic effect of gemcitabine on pancreatic cancer cells sistance in TP53- or ATM-defective chronic lymphocytic leukemia cells.
through enhancement of DNA damage and abrogation of ribonucleotide Blood 2016;127:582–95.
reductase induction by gemcitabine. Oncol Rep 2017;37:3377–86. 79. Higgins GS, Boulton SJ. Beyond PARP—POLq as an anticancer target.
58. Wallez Y, Dunlop CR, Johnson TI, Koh S-B, Fornari C, Yates JWT, et al. The Science 2018;359:1217–8.
ATR inhibitor AZD6738 synergizes with gemcitabine in vitro and in vivo to 80. Leon TE, Rapoz-D'Silva T, Rahman S, Magnussen M, Farah N, Guerra-
induce pancreatic ductal adenocarcinoma regression. Mol Cancer Ther Assunç~ao JA, et al. Synthetic lethal screening identifies CHK1 inhibition as
2018;17:1670–82. an exploitable vulnerability in EZH2 deficient T-ALL. Blood 2017;130:
59. Koh SB, Courtin A, Boyce RJ, Boyle RG, Richards FM, Jodrell DI. CHK1 1325.
inhibition synergizes with gemcitabine initially by destabilizing the DNA 81. Dietlein F, Kalb B, Jokic M, Noll EM, Strong A, Tharun L, et al. A synergistic
replication apparatus. Cancer Res 2015;75:3583–95. interaction between Chk1- and MK2 Inhibitors in KRAS-mutant cancer.
60. Liu Y, Li Y, Wang X, Liu F, Gao P, Quinn MM, et al. Gemcitabine and Chk1 Cell 2015;162:146–59.
inhibitor AZD7762 synergistically suppress the growth of Lkb1-deficient 82. Pfister SX, Markkanen E, Jiang Y, Sarkar S, Woodcock M, Orlando G, et al.
lung adenocarcinoma. Cancer Res 2017;77:5068–76. Inhibiting WEE1 selectively kills histone H3K36me3-deficient cancers by
61. Vendetti FP, Lau A, Schamus S, Conrads TP, O'Connor MJ, Bakkenist CJ. dNTP starvation. Cancer Cell 2015;28:557–68.
The orally active and bioavailable ATR kinase inhibitor AZD6738 potenti- 83. Kotsantis P, Petermann E, Boulton SJ. Mechanisms of oncogene-
ates the anti-tumor effects of cisplatin to resolve ATM-deficient non-small induced replication stress: Jigsaw falling into place. Cancer Discov
cell lung cancer in vivo. Oncotarget 2015;6:44289–305. 2018;8:537–55.
62. Li CC, Yang JC, Lu MC, Lee CL, Peng CY, Hsu WY, et al. ATR-Chk1 84. Toledo LI, Murga M, Zur R, Soria R, Rodriguez A, Martinez S, et al. A cell-
signaling inhibition as a therapeutic strategy to enhance cisplatin che- based screen identifies ATR inhibitors with synthetic lethal properties for
mosensitivity in urothelial bladder cancer. Oncotarget 2016;7:1947–59. cancer-associated mutations. Nat Struct Mol Biol 2011;18:721–7.
63. Josse R, Martin SE, Guha R, Ormanoglu P, Pfister TD, Reaper PM, et al. ATR 85. Schoppy DW, Ragland RL, Gilad O, Shastri N, Peters AA, Murga M, et al.
inhibitors VE-821 and VX-970 sensitize cancer cells to topoisomerase I Oncogenic stress sensitizes murine cancers to hypomorphic suppression
inhibitors by disabling DNA replication initiation and fork elongation of ATR. J Clin Invest 2012;122:241–52.
responses. Cancer Res 2014;74:6968–79. 86. Gilad O, Nabet BY, Ragland RL, Schoppy DW, Smith KD, Durham AC,
64. Flatten K, Dai NT, Vroman BT, Loegering D, Erlichman C, Karnitz LM, et al. et al. Combining ATR suppression with oncogenic Ras synergistically
The role of checkpoint kinase 1 in sensitivity to topoisomerase I poisons. increases genomic instability, causing synthetic lethality or tumorigenesis
J Biol Chem 2005;280:14349–55. in a dosage-dependent manner. Cancer Res 2010;70:9693–702.
65. Hong D, Infante J, Janku F, Jones S, Nguyen LM, Burris H, et al. Phase I 87. Sanjiv K, Hagenkort A, Calder on-Monta~ no JM, Koolmeister T, Reaper PM,
study of LY2606368, a checkpoint kinase 1 inhibitor, in patients with Mortusewicz O, et al. Cancer-specific synthetic lethality between ATR and
advanced cancer. J Clin Oncol 2016;34:1764–71. CHK1 kinase activities. Cell Rep 2016;14:298–309.
66. Buisson R, Boisvert JL, Benes CH, Zou L. Distinct but concerted roles of 88. Nikkil€a J, Kumar R, Campbell J, Brandsma I, Pemberton HN, Wallberg F,
ATR, DNA-PK, and Chk1 in countering replication stress during S phase. et al. Elevated APOBEC3B expression drives a kataegic-like mutation
Mol Cell 2015;59:1011–24. signature and replication stress-related therapeutic vulnerabilities in
67. Leijen S, van Geel RM, Pavlick AC, Tibes R, Rosen L, Razak AR, et al. Phase I p53-defective cells. Br J Cancer 2017;117:113–23.
study evaluating WEE1 inhibitor AZD1775 as monotherapy and in 89. Buisson R, Lawrence MS, Benes CH, Zou L. APOBEC3A and APOBEC3B
combination with gemcitabine, cisplatin, or carboplatin in patients with activities render cancer cells susceptible to ATR inhibition. Cancer Res
advanced solid tumors. J Clin Oncol 2016;34:4371–80. 2017;77:4567–78.
68. De Witt Hamer PC, Mir SE, Noske D, Van Noorden CJ, W€ urdinger T. WEE1 90. Burns MB, Temiz NA, Harris RS. Evidence for APOBEC3B mutagenesis in
kinase targeting combined with DNA-damaging cancer therapy catalyzes multiple human cancers. Nat Genet 2013;45:977–83.
mitotic catastrophe. Clin Cancer Res 2011;17:4200–7. 91. Roberts SA, Lawrence MS, Klimczak LJ, Grimm SA, Fargo D, Stojanov P,
69. Domínguez-Kelly R, Martín Y, Koundrioukoff S, Tanenbaum ME, Smits et al. An APOBEC cytidine deaminase mutagenesis pattern is widespread
VA, Medema RH, et al. Wee1 controls genomic stability during replication in human cancers. Nat Genet 2013;45:970–6.
by regulating the Mus81-Eme1 endonuclease. J Cell Biol 2011;194: 92. Luthra P, Aguirre S, Yen BC, Pietzsch CA, Sanchez-Aparicio MT, Tigabu B,
567–79. et al. Topoisomerase II inhibitors induce DNA damage-dependent
70. Beck H, Nahse-Kumpf V, Larsen MS, O'Hanlon KA, Patzke S, Holmberg C, interferon responses circumventing ebola virus immune evasion. MBio
et al. Cyclin-dependent kinase suppression by WEE1 kinase protects the 2017;8:pii: e00368–17.

1738 Cancer Res; 79(8) April 15, 2019 Cancer Research


Exploiting Replication Stress

93. Coquel F, Silva MJ, Techer H, Zadorozhny K, Sharma S, Nieminuszczy J, 103. Xia T, Konno H, Ahn J, Barber GN. Deregulation of STING signaling in
et al. SAMHD1 acts at stalled replication forks to prevent interferon colorectal carcinoma constrains DNA damage responses and correlates
induction. Nature 2018;557:57–61. with tumorigenesis. Cell Rep 2016;14:282–97.
94. Dunphy G, Flannery SM, Almine JF, Connolly DJ, Paulus C, Jønsson KL, 104. Somyajit K, Gupta R, Sedlackova H, Neelsen KJ, Ochs F, Rask MB, et al.
et al. Non-canonical activation of the DNA sensing adaptor STING by Redox-sensitive alteration of replisome architecture safeguards genome
ATM and IFI16 mediates NF-kB signaling after nuclear DNA damage. integrity. Science 2017;358:797–802.
Mol Cell 2018;71:745–60. 105. Toledo CM, Ding Y, Hoellerbauer P, Davis RJ, Basom R, Girard EJ, et al.
95. Shen J, Zhao W, Ju Z, Wang L, Peng G. PARPi triggers STING-dependent Genome-wide CRISPR-Cas9 screens reveal loss of redundancy between
immune response and enhances therapeutic efficacy of immune check- PKMYT1 and WEE1 in glioblastoma stem-like cells. Cell Rep 2015;13:
point blockade independent of BRCAness. Cancer Res 2018;79:311–9. 2425–39.
96. Harding SM, Benci JL, Irianto J, Discher DE, Minn AJ, Greenberg RA. 106. Wang T, Yu H, Hughes NW, Liu B, Kendirli A, Klein K, et al. Gene
Mitotic progression following DNA damage enables pattern recognition essentiality profiling reveals gene networks and synthetic lethal interac-
within micronuclei. Nature 2017;548:466–70. tions with oncogenic Ras. Cell 2017;168:890–903.
97. Mackenzie KJ, Carroll P, Martin CA, Murina O, Fluteau A, Simpson DJ, 107. Kanarek N, Keys HR, Cantor JR, Lewis CA, Chan SH, Kunchok T, et al.
et al. cGAS surveillance of micronuclei links genome instability to innate Histidine catabolism is a major determinant of methotrexate sensitivity.
immunity. Nature 2017;548:461–5. Nature 2018;559:632–6.
98. Bhattacharya S, Srinivasan K, Abdisalaam S, Su F, Raj P, Dozmorov I, et al. 108. Zimmermann M, Murina O, Reijns MAM, Agathanggelou A, Challis
RAD51 interconnects between DNA replication, DNA repair and immu- R, Tarnauskaite Z,  et al. CRISPR screens identify genomic ribonu-
nity. Nucleic Acids Res 2017;45:4590–605. cleotides as a source of PARP-trapping lesions. Nature 2018;559:
99. Erdal E, Haider S, Rehwinkel J, Harris AL, McHugh PJ. A prosurvival DNA 285–9.

Downloaded from http://aacrjournals.org/cancerres/article-pdf/79/8/1730/2791709/1730.pdf by guest on 05 March 2023


damage-induced cytoplasmic interferon response is mediated by end 109. Jordheim LP, Durantel D, Zoulim F, Dumontet C. Advances in the
resection factors and is limited by Trex1. Genes Dev 2017;31:353–69. development of nucleoside and nucleotide analogues for cancer and
100. Shen YJ, Le Bert N, Chitre AA, Koo CX, Nga XH, Ho SS, et al. Genome- viral diseases. Nat Rev Drug Discov 2013;12:447–64.
derived cytosolic DNA mediates type I interferon-dependent rejection of 110. Lord CJ, Ashworth A. PARP inhibitors: synthetic lethality in the clinic.
B cell lymphoma cells. Cell Rep 2015;11:460–73. Science 2017;355:1152–8.
101. Wolf C, Rapp A, Berndt N, Staroske W, Schuster M, Dobrick-Mattheuer M, 111. Ma CX, Janetka JW, Piwnica-Worms H. Death by releasing the breaks:
et al. RPA and Rad51 constitute a cell intrinsic mechanism to protect the CHK1 inhibitors as cancer therapeutics. Trends Mol Med 2011;17:
cytosol from self DNA. Nat Commun 2016;7:11752. 88–96.
102. Li T, Chen ZJ. The cGAS–cGAMP–STING pathway connects DNA damage 112. Matheson CJ, Backos DS, Reigan P. Targeting WEE1 kinase in cancer.
to inflammation, senescence, and cancer. J Exp Med 2018;215:1287–99. Trends Pharmacol Sci 2016;37:872–81.

www.aacrjournals.org Cancer Res; 79(8) April 15, 2019 1739

You might also like