Inhibitors of various elements of the DNA repair pathways have entered clinical development or are in late preclinical stages of drug development. It was initially considered that agents targeting DNA repair would act to overcome tumor resistance to chemotherapy and radiotherapy. More recent data have shown that targeting DNA repair pathways can be effective in selected tumors via a synthetically lethal route, with single agent activity having been shown with poly-ADP ribose polymerase (PARP) inhibitors. An increased understanding of the biology and interaction of the DNA repair pathways also means that rational combination of DNA repair inhibitors may also give great benefit in the clinic. Clin Cancer Res; 16(18); 4527–31. ©2010 AACR.

Repair of DNA and, thus, preservation of the genetic code are critical for normal cellular function. To this end, human cells have at least five recognized pathways that protect the genome by signaling specific types of DNA damage and carrying out repair (reviewed in refs. 13). In cancer cells, these pathways represent a curious dichotomy; it is well recognized that mutations in the pathways can predispose to cancer and are hallmarks of many of the hereditary cancer syndromes (46). However, once an immortalized tumor cell has developed, the DNA repair pathways can be used by this cell to overcome many of our standard anticancer treatments and, hence, are a cause of treatment resistance. Increasing evidence in the literature shows that tumor tissue has high levels of some elements of the DNA repair pathways (7, 8), and is able to use these pathways to repair damage caused by many of our standard anticancer therapies. Therefore, inhibiting DNA repair may “level the playing field” and make the tumor more vulnerable to treatment.

The major DNA repair pathways are direct repair, mismatch repair (MMR), base excision repair (BER), nucleotide excision repair (NER), and double-strand break (DSB) recombinational repair, which includes both nonhomologous end-joining (NHEJ) and homologous recombinational repair (HHR; refs. 1, 2, 9). O6-alkylguanine-DNA alkyltransferase (MGMT, OGAT, ATase) is the main component of the direct repair pathway, an efficient mechanism of DNA repair in which the altered base is corrected without removal or disruption of the phosphodiester backbone. Overexpression of ATase in mammalian cells confers resistance to DNA-alkylating agents (reviewed in ref. 10), and is a major factor in tumor resistance to these drugs. NER is involved in the repair of UV damage and removal of bulky DNA adducts, such as those caused by cross-linking agents. MMR repairs replication errors and is frequently mutated in cancer cells allowing tolerance of such lesions (1113). BER is involved in the repair of single-strand breaks, contributing to resistance to ionizing radiation and alkylating agents. Recombinational repair has two pathways: the error-free HRR in dividing cells and error-prone NHEJ active in G1. These two pathways repair much of the damage caused by radiotherapy and chemotherapeutic agents, such as cisplatin and mitomycin C (2).

Some compounds in the clinic or in late preclinical development inhibit direct repair and elements of the base excision and DSB repair pathways. The initial development of inhibitors of DNA repair pathways was designed to overcome chemo- or radio-resistance (1419). However, the increasing knowledge about the complexity and interactions of the DNA damage response pathways, as well as the entry of a range of compounds into the clinic, have led to a fascinating area of drug development, with opportunities for improving on existing treatments and for the design of rational combinations of novel agents to improve treatment response. Table 1 shows the range and variety of these DNA damage response modulators that have entered the clinic in recent years.

Table 1.

DNA damage response–modulating drugs in clinical development, grouped by repair pathway targeted

AgentCompanyAdministrationSingle and/or Combination TherapyDisease indicationsClinical status
Direct repair (MGMT) 
O6benzylguanine   Combination BCNU GBM Phase II complete 
Lomeguatrib KuDos Oral Combination with TMZ Melanoma Phase II complete 
 
Single-strand break repair (PARP inhibitors, PARPi) 
PF0367338 (AG014699) Pfizer IV Combination ++ single agent Solid tumors, melanoma Phase I and II complete and others ongoing 
Olaparib (AZD2281) AztraZeneca (KuDos) Oral Combination ++ single agent BRCA defective, solid tumors various Phase I and II studies completed and ongoing 
Veliparib (ABT888) Abbott Oral Combination ++ Various solid tumors Phase I and II studies completed and ongoing 
Iniparib (SAR240550, BSI 201) Sanofi Aventis (Bipar) IV Combination Triple negative breast Phase III complete 
MK4827 Merck Oral Single agent Solid, BRCA ovarian Phase I ongoing 
CEP-9722 Cephalon Oral Combination with TMZ Solid tumors Phase I ongoing 
E7016 (GPI 21016) Eisai (MGI Pharma) Oral Combination with TMZ Solid tumors Phase I ongoing 
LT673 Biomarin Oral  Solid tumors Phase I planned 
 
Single-strand break repair (APE1 inhibitors APE1i) 
TRC102 Tracon Oral Combination with pemetrexed Solid tumors Phase I complete 
Methoxyamine  IV Combination with TMZ Solid tumors Phase I suspended 
 
DSB repair (RAD51 inhibitor, RAD51i) 
MP470 Supergen Oral Single agent Lymphoma and/or solid tumors Phase I ? opened 
 
DSB repair (ATM inhibitor, ATMi) 
KU55933 AstraZeneca (KuDos) Preclinical 
CP466722 Pfizer Preclinical 
 
DSB repair (DNA PK inhibitor, DNA PKi) 
NU7441 AstraZeneca (KuDos) Preclinical 
AgentCompanyAdministrationSingle and/or Combination TherapyDisease indicationsClinical status
Direct repair (MGMT) 
O6benzylguanine   Combination BCNU GBM Phase II complete 
Lomeguatrib KuDos Oral Combination with TMZ Melanoma Phase II complete 
 
Single-strand break repair (PARP inhibitors, PARPi) 
PF0367338 (AG014699) Pfizer IV Combination ++ single agent Solid tumors, melanoma Phase I and II complete and others ongoing 
Olaparib (AZD2281) AztraZeneca (KuDos) Oral Combination ++ single agent BRCA defective, solid tumors various Phase I and II studies completed and ongoing 
Veliparib (ABT888) Abbott Oral Combination ++ Various solid tumors Phase I and II studies completed and ongoing 
Iniparib (SAR240550, BSI 201) Sanofi Aventis (Bipar) IV Combination Triple negative breast Phase III complete 
MK4827 Merck Oral Single agent Solid, BRCA ovarian Phase I ongoing 
CEP-9722 Cephalon Oral Combination with TMZ Solid tumors Phase I ongoing 
E7016 (GPI 21016) Eisai (MGI Pharma) Oral Combination with TMZ Solid tumors Phase I ongoing 
LT673 Biomarin Oral  Solid tumors Phase I planned 
 
Single-strand break repair (APE1 inhibitors APE1i) 
TRC102 Tracon Oral Combination with pemetrexed Solid tumors Phase I complete 
Methoxyamine  IV Combination with TMZ Solid tumors Phase I suspended 
 
DSB repair (RAD51 inhibitor, RAD51i) 
MP470 Supergen Oral Single agent Lymphoma and/or solid tumors Phase I ? opened 
 
DSB repair (ATM inhibitor, ATMi) 
KU55933 AstraZeneca (KuDos) Preclinical 
CP466722 Pfizer Preclinical 
 
DSB repair (DNA PK inhibitor, DNA PKi) 
NU7441 AstraZeneca (KuDos) Preclinical 

NOTE. Data taken from ClinicalTrials.gov (http://clinicaltrials.gov/) compounds that are thought to be in late preclinical development are also included for completeness.

Abbreviations: IV, intravenous; TMZ, temozolomide; GBM, glioblastoma multiforme; BCNU, carmustine or bis-xhloronitrosourea; ? indicates clinical studies not yet initiated.

When considering the pipeline of novel agents that have entered clinical development or are at a late preclinical phase over the last 5 years, it is worth briefly reviewing the earlier trials in which blocking a DNA repair pathway was the primary aim. To date, a common theme has emerged in the majority of trials, which seems to be limiting the effectiveness of this strategy potentiation of normal tissue toxicity. Depletion of MGMT and, hence, disruption of the direct repair pathway by O6benzylguanine or lomeguatrib was successfully achieved more than 10 years ago. These agents were combined with carmustine and temozolomide, respectively, and although pharmacodynamic assays confirmed depletion of the target, this was achieved at the expense of the chemotherapeutic dose (14, 17). Enhanced normal tissue toxicity, in the form of more profound myelosuppression, meant that a significant reduction in chemotherapy dose was required, and phase II studies did not show a benefit in terms of increased tumor response (15, 18). When the first poly-ADP ribose polymerase (PARP) inhibitor in cancer treatment entered the clinic in 2003, it was also evaluated in combination with chemotherapy, with the initial reports being that a PARP-inhibitory dose of drug could be given with full dose temozolomide (19). This finding was not borne out in the subsequent phase II study, in which a 25% reduction in cytotoxic dose was needed for a tolerable regimen (20). Although this small study did suggest a possible clinical benefit of the combination, this has yet to be confirmed in a randomized study.

Enhancement of normal tissue toxicity is emerging as a common theme with some of the other PARP inhibitors when combined with chemotherapy. Studies with ABT888 (veliparib) and AZD2281 (olaparib) with a range of cytotoxic agents have reported the need to reduce chemotherapy dose because of enhanced myelosuppression (2123). The outlying data in this area are from the combination of BSI-201 with carboplatin and gemcitabine in triple negative breast cancer, in which very encouraging evidence of increased activity was observed with no increase in toxicity (24, 25). It has been speculated that this may be due to the intermittent schedule of dosing of the PARP inhibitor allowing bone marrow recovery. This theory would argue for the increased activity being due to chemo-potentiation, rather than single-agent PARP activity, acting through synthetic lethality on the proposed HRR-deficient triple-negative phenotype, as other studies showing single-agent activity have required continuous and profound PARP inhibition (2628). Other agents targeting DNA damage response in late preclinical development are entering the clinic [inhibitors of DNA-dependent protein kinase (DNA PK), ataxia telangiectasia mutated (ATM), ATM- and Rad3-related (ATR), and RAD51; refs. 2933]. These agents have also shown the ability to potentiate the activity of cytotoxic drugs in preclinical models; it remains to be seen whether this can be done without the increased toxicity and subsequent dose reductions that have been required in many of the previous studies. It may be that we are able to use these powerful inhibitors to fuller potential in the area of radio-potentiation. Radiation causes DNA single- and double-strand breaks, and many of the DNA repair inhibitors have been shown to be radio-potentiating (29, 30, 34). The increasing use of highly technical radiotherapy techniques [intensity modulated radiation therapy (IMRT), image-guided radiation therapy (IGRT), and tomotherapy] may allow radiation and/or inhibitor combination studies in which tumor response is improved without consequent increase in normal tissue toxicity.

One of the very exciting developments in the field of DNA damage response research in the last few years has been the preclinical (35, 36) and subsequent clinical demonstration (2628) of the ability to cause synthetic lethality in selected cell types using a DNA repair inhibitor without also using a DNA damaging agent. Although, first shown with the use of PARP inhibitors in patients with familial breast and ovarian cancer carrying the BRCA genes, it has opened up the possibility that this much less toxic strategy may be a benefit in patients with sporadic tumors if a predictive molecular phenotype can be identified. Many research groups are now working to develop functional assays for DSB repair competence, or molecular signatures that will allow enrichment of patient populations within trials. An additional consequence of this pioneering research has been that DNA repair inhibitors have been recognized as potentially active anticancer agents in their own right. With the expanding knowledge of the DNA damage response pathways and the plethora of drugs entering clinical development targeting different elements of these pathways, it will be possible to design trials in which novel combinations of repair inhibitors, including the checkpoint inhibitors (3739), may be active, or in which patients are selected on the basis of the oncogenic mutation status of their tumor, for example mutations in ATR, ATM, or the Fanconi proteins may also predict for sensitivity to PARP inhibitors (40, 41). Figure 1A summarizes, in simplified form, the BER and DSB pathways, emphasizing the close interactions and cross talk between the BER and DSB repair pathways and cell-cycle checkpoint signaling. Figure 1B illustrates the current DNA damage response–modulating drugs targeting these pathways and related checkpoint signaling illustrating the fascinating potential for rational combination of these novel agents.

Fig. 1.

Simplified schematic of signaling and repair of single- and double-DNA strand breaks to illustrate the interactions between pathways and potential rational inhibitor combinations. A, damaged DNA bases are excised involving APE-1, and single-strand break (SSB) activates PARP, which recruits other components of BER. In a dividing cell, unrepaired SSB will become DSB. The presence of DSBs is signaled via the ATM/ATR pathways. In nondividing cells, G1 arrest is signaled via Chk2, and NHEJ pathways repair the break. In dividing cells, G2 arrest allows error-free repair using HR. B, simplified schematic of signaling and repair of single- and double-DNA strand breaks showing the points of action of current inhibitors in the clinic or in late preclinical development. Additionally, it is highlighted that Chk1 inhibitors have entered the clinic, presenting an opportunity to combine these agents with DNA damage response modulators.

Fig. 1.

Simplified schematic of signaling and repair of single- and double-DNA strand breaks to illustrate the interactions between pathways and potential rational inhibitor combinations. A, damaged DNA bases are excised involving APE-1, and single-strand break (SSB) activates PARP, which recruits other components of BER. In a dividing cell, unrepaired SSB will become DSB. The presence of DSBs is signaled via the ATM/ATR pathways. In nondividing cells, G1 arrest is signaled via Chk2, and NHEJ pathways repair the break. In dividing cells, G2 arrest allows error-free repair using HR. B, simplified schematic of signaling and repair of single- and double-DNA strand breaks showing the points of action of current inhibitors in the clinic or in late preclinical development. Additionally, it is highlighted that Chk1 inhibitors have entered the clinic, presenting an opportunity to combine these agents with DNA damage response modulators.

Close modal

As the DNA repair inhibitors continue to move forward in clinical development, we need to be able to learn from and build on the lessons of history, so that the true potential of these drugs is realized. It is likely that they will ultimately be used in combination regimens; the response rates in the BRCA population are not at the levels seen in chronic myeloid leukemia and gastrointestinal stromal tumor with imatinib. Therefore, much work remains to be done in exploring scheduling to avoid increased toxicities. When a DNA damage–modulating agent is to be used to prevent repair, and so potentiate the activity of the cytotoxic agent, the drugs need to be given concurrently. It may be that pulsed schedules of the modulator would, in this situation, cause the desired tumor-cell kill, but allow normal tissue toxicity to recover. If the modulator is predicted to have single-agent activity in a particular disease setting, then scheduling apart from the cytotoxic with longer duration of coverage might be the optimal route. The interplay between the increasing knowledge of the biology of these pathways and the increasing ability to explore the molecular profile of our patients' tumors using array- and circulating-tumor cell technologies means that this is an exciting, fast moving, and potentially very beneficial area of cancer treatment research.

R. Plummer, ownership interest and commercial research support for AG014699.

1
Hoeijmakers
JH
. 
Genone maintenance mechanisms for preventing cancer
.
Nature
2001
;
411
:
366
74
.
2
Bernstein
C
,
Bernstein
H
,
Payne
CM
,
Garewal
H
. 
DNA repair/pro-apoptotic dual-role proteins in five major DNA repair pathways: fail-safe protection against carcinogenesis
.
Mutat Res
2002
;
511
:
145
78
.
3
Christmann
M
,
Tomicic
MT
,
Roos
WP
,
Kaina
B
. 
Mechanisms of human DNA repair: an update
.
Toxicology
2003
;
193
:
3
34
.
4
Heinen
CD
,
Schmutte
C
,
Fishel
R
. 
DNA repair and tumorigenesis: lessons from hereditary cancer syndromes
.
Cancer Biol Ther
2002
;
1
:
477
85
.
5
Risinger
MA
,
Groden
J
. 
Crosslinks and crosstalk: human cancer syndromes and DNA repair defects
.
Cancer Cell
2004
;
6
:
539
45
.
6
de la Chapelle
A
. 
Genetic predisposition to colorectal cancer
.
Nat Rev Cancer
2004
;
4
:
769
80
.
7
Staibano
S
,
Pepe
S
,
Lo Muzio
L
, et al
. 
Poly(adenosine diphosphate-ribose) polymerase 1 expression in malignant melanomas from photoexposed areas of the head and neck region
.
Hum Pathol
2005
;
36
:
724
31
.
8
Wharton
SB
,
McNelis
U
,
Bell
HS
,
Whittle
IR
. 
Expression of poly(ADP-ribose) polymerase and distribution of poly(ADP-ribosyl)ation in glioblastoma and in a glioma multicellular tumour spheroid model
.
Neuropathol Appl Neurobiol
2000
;
26
:
528
35
.
9
Hansen
WK
,
Kelly
M
. 
Review of mammalian DNA repair and translational implications
.
J Pharmacol Exp Ther
2000
;
295
:
1
9
.
10
Margison
GP
,
Santibáñez Koref
MS
,
Povey
AC
. 
Mechanisms of carcinogenicity/chemotherapy by O6-methylguanine
.
Mutagenesis
2002
;
17
:
483
7
.
11
Fink
D
,
Aebi
S
,
Howell
S
. 
The role of DNA mismatch repair in drug resistance
.
Clin Cancer Res
1998
;
4
:
1
6
.
12
Soejima
H
,
Zhao
W
,
Mukai
T
. 
Epigenetic silencing of the MGMT gene in cancer
.
Biochem Cell Biol
2005
;
83
:
429
37
.
13
Park
Y
,
Gerson
SL
. 
DNA repair defects in stem cell function and aging
.
Annu Rev Med
2005
;
56
:
495
508
.
14
Schilsky
RL
,
Dolan
ME
,
Bertucci
D
, et al
. 
Phase I clinical and pharmacological study of O6-benzylguanine followed by carmustine in patients with advanced cancer
.
Clin Cancer Res
2000
;
6
:
3025
31
.
15
Quinn
JA
,
Pluda
J
,
Dolan
ME
, et al
. 
Phase II trial of carmustine plus O(6)-benzylguanine for patients with nitrosourea-resistant recurrent or progressive malignant glioma
.
J Clin Oncol
2002
;
20
:
2277
83
.
16
Gajewski
TF
,
Sosman
J
,
Gerson
SL
, et al
. 
Phase II trial of the O6-alkylguanine DNA alkyltransferase inhibitor O6-benzylguanine and 1,3-bis(2-chloroethyl)-1-nitrosourea in advanced melanoma
.
Clin Cancer Res
2005
;
11
:
7861
5
.
17
Ranson
M
,
Middleton
MR
,
Bridgewater
J
, et al
. 
Lomeguatrib, a potent inhibitor of O6-alkylguanine-DNA-alkyltransferase: phase I safety, pharmacodynamic, and pharmacokinetic trial and evaluation in combination with temozolomide in patients with advanced solid tumors
.
Clin Cancer Res
2006
;
12
:
1577
84
.
18
Ranson
M
,
Hersey
P
,
Thompson
D
, et al
. 
A randomised trial of the combination of lomeguatrib and temozolomide alone in patients with advanced melanoma
.
J Clin Oncol
2007
;
25
:
2540
5
.
19
Plummer
R
,
Jones
C
,
Middleton
M
, et al
. 
Phase I study of the poly(ADP-Ribose) polymerase inhibitor, AG014699, in combination with temozolomide in patients with advanced solid tumors
.
Clin Cancer Res
2008
;
14
:
7917
23
.
20
Plummer
R
,
Lorigan
P
,
Evans
J
, et al
. 
First and final report of a phase II study of the poly(ADP-ribose) polymerase (PARP) inhibitor, AG014699, in combination with temozolomide (TMZ) in patients with metastatic malignant melanoma (MM)
.
J Clin Oncol
2006
;
24
:
8013
.
21
Rajan
A
,
Carter
CA
,
Gutierrez
M
, et al
. 
A phase I combination study of olaparib (AZD2281; KU-0059436) and cisplatin plus gemcitabine in adults with solid tumors
.
J Thorac Oncol
2009
;
4
:
S598
9
.
22
Rajan
A
,
Gutierrez
M
,
Kummar
S
, et al
. 
A phase I combination study of Azd2281 and cisplatin plus gemcitabine in adults with solid tumors
.
Ann Oncol
2009
;
20
:
42
3
.
23
Kummar
S
,
Ji
J
,
Zhang
Y
, et al
. 
A phase I combination study of Abt-888 and topotecan hydrochloride in adults with refractory solid tumors and lymphomas
.
Ann Oncol
2009
;
20
:
42
.
24
O'Shaughnessy
J
,
Osborne
C
,
Pippen
J
, et al
. 
Efficacy of BSI-201, a poly (ADP-ribose) polymerase-1 (PARP1) inhibitor, in combination with gemcitabine/carboplatin (G/C) in patients with metastatic triple-negative breast cancer (TNBC): Results of a randomized phase II trial
.
J Clin Oncol
2009
;
27
:
3
.
25
BiPar Sciences presents interim phase 2 results for PARP inhibitor BSI-201 at San Antonio Breast Cancer Symposium
.
Cancer Biol Ther
2009
;
8
:
2
3
.
26
Audeh
M
,
Penson
R
,
Friedlander
M
, et al
. 
Phase II trial of the oral PARP inhibitor olaparib (AZD2281) in BRCA-deficient advanced ovarian cancer
.
J Clin Oncol
2009
;
27
:
5500
.
27
Tutt
A
,
Robson
M
,
Garber
J
, et al
. 
Phase II trial of the oral PARP inhibitor olaparib in BRCA-deficient advanced breast cancer
.
J Clin Oncol
2009
;
27
:
CRA501
.
28
Fong
P
,
Boss
D
,
Yap
T
, et al
. 
Inhibition of poly(ADP-Ribose) polymerase in tumors from BRCA mutation carriers
.
N Engl J Med
2009
;
361
:
123
34
.
29
Griffin
RJ
,
Fontana
G
,
Golding
BT
, et al
. 
Selective benzopyranone and pyrimido[2,1-a]isoquinolin-4-one inhibitors of DNA-dependent protein kinase: synthesis, structure-activity studies, and radiosensitization of a human tumor cell line in vitro
.
J Med Chem
2005
;
48
:
569
85
.
30
Hickson
I
,
Zhao
Y
,
Richardson
CJ
, et al
. 
Identification and characterization of a novel and specific inhibitor of the ataxia-telangiectasia mutated kinase ATM
.
Cancer Res
2004
;
64
:
9152
9
.
31
Huang
G
,
Wang
H
,
Yang
LX
, et al
. 
Enhancement of radiation-induced DNA damage and inhibition of its repair by a novel camptothecin analog
.
Anticancer Res
2009
;
30
:
937
44
.
32
Ismail
IH
,
Martensson
S
,
Moshinsky
D
, et al
. 
SU11752 inhibits the DNA-dependent protein kinase and DNA double-strand break repair resulting in ionizing radiation sensitization
.
Oncogene
2004
;
23
:
873
82
.
33
Hollick
JJ
,
Rigoreau
LJ
,
Cano-Soumillac
C
, et al
. 
Pyranone, thiopyranone, and pyridone inhibitors of phosphatidylinositol 3-kinase related kinases. Structure-activity relationships for DNA-dependent protein kinase inhibition, and identification of the first potent and selective inhibitor of the ataxia telangiectasia mutated kinase
.
J Med Chem
2007
;
50
:
1958
72
.
34
Calabrese
CR
,
Almassy
R
,
Barton
S
, et al
. 
Anticancer chemosensitization and radiosensitization by the novel poly(ADP-ribose) polymerase-1 inhibitor AG14361
.
J Natl Cancer Inst
2004
;
96
:
56
67
.
35
Bryant
HE
,
Schultz
N
,
Thomas
HD
, et al
. 
Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase
.
Nature
2005
;
434
:
913
7
.
36
Farmer
H
,
McCabe
N
,
Lord
CJ
, et al
. 
Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy
.
Nature
2005
;
434
:
917
21
.
37
Ashwell
S
,
Janetka
JW
,
Zabludoff
S
. 
Keeping checkpoint kinases in line: new selective inhibitors in clinical trials
.
Expert Opin Investig Drugs
2008
;
17
:
1331
40
.
38
Janetka
JW
,
Ashwell
S
. 
Checkpoint kinase inhibitors: a review of the patent literature
.
Expert Opin Ther Pat
2009
;
19
:
165
97
.
39
Kuntz
K
,
O'Connell
MJ
. 
The G(2) DNA damage checkpoint: could this ancient regulator be the Achilles heel of cancer?
Cancer Biol Ther
2009
;
8
:
1433
9
.
40
Kennedy
RD
,
D'Andrea
AD
. 
DNA repair pathways in clinical practice: lessons from pediatric cancer susceptibility syndromes
.
J Clin Oncol
2006
;
24
:
3799
808
.
41
Kennedy
RD
,
Chen
CC
,
Stuckert
P
, et al
. 
Fanconi anemia pathway-deficient tumour cells are hypersensitive to inhibition of ataxia telangiectasia mutated
.
J Clin Invest
2007
;
117
:
1440
9
.