Purpose:

The ATM (ataxia telangiectasia mutated) gene is mutated in a subset of prostate cancers, and ATM mutation may confer specific therapeutic vulnerabilities, although ATM-deficient prostate cancers have not been well-characterized.

Experimental Design:

We genetically validated a clinical grade IHC assay to detect ATM protein loss and examined the frequency of ATM loss among tumors with pathogenic germline ATM mutations and genetically unselected primary prostate carcinomas using tissue microarrays (TMAs). Immunostaining results were correlated with targeted somatic genomic sequencing and clinical outcomes.

Results:

ATM protein loss was found in 13% (7/52) of primary Gleason pattern 5 cancers with available sequencing data and was 100% sensitive for biallelic ATM inactivation. In a separate cohort with pathogenic germline ATM mutations, 74% (14/19) had ATM protein loss of which 70% (7/10) of evaluable cases had genomic evidence of biallelic inactivation, compared with zero of four of cases with intact ATM expression. By TMA screening, ATM loss was identified in 3% (25/831) of evaluable primary tumors, more commonly in grade group 5 (17/181; 9%) compared with all other grades (8/650; 1%; P < 0.0001). Of those with available sequencing, 80% (4/5) with homogeneous ATM protein loss and 50% (6/12) with heterogeneous ATM protein loss had detectable pathogenic ATM alterations. In surgically treated patients, ATM loss was not significantly associated with clinical outcomes in random-effects Cox models after adjusting for clinicopathologic variables.

Conclusions:

ATM loss is enriched among high-grade prostate cancers. Optimal evaluation of ATM status requires both genomic and IHC studies and will guide development of molecularly targeted therapies.

Translational Relevance

Given the key role of ATM (ataxia telangiectasia mutated) as a signal transducer of double-stranded DNA breaks, prostate cancers with ATM inactivation have largely been assumed to be similar to those with BRCA1/2 alterations. However, emerging data suggest that ATM-deficient tumors may be biologically distinct and derive less benefit from PARP inhibitors and potentially more benefit from other targeted therapies compared with BRCA1/2-mutated prostate cancers. Here, we develop and genetically validate a clinical grade IHC assay to detect ATM loss and use this assay to identify and characterize several cohorts of ATM-deficient prostate cancers. We demonstrate that ATM loss is highly enriched among high-grade prostate cancers and find that loss is frequently focal and a relatively late molecular event. We propose a combined interrogation approach utilizing both ATM IHC and next-generation sequencing assays to identify ATM-deficient tumors which may benefit from future trials of targeted therapies.

The ataxia telangiectasia mutated (ATM) gene encodes a PI3K-related serine/threonine kinase involved in the maintenance of genomic integrity. ATM plays a central role in the sensing and cellular response to DNA damage, and acts as a key signal transducer in the double-strand break repair process (1). Biallelic germline mutation of the ATM gene leads to ataxia telangiectasia syndrome, characterized by neurodegeneration, immune deficiencies, and increased cancer susceptibility (2). Heterozygous deleterious germline mutations in ATM occur in around 0.5%–1% of the population and are also associated with tumor predisposition, including breast, pancreatic, lung, thyroid, and prostate cancers (2). In prostate cancer, tumoral ATM mutations may be either germline or somatic in origin, and are present in 5%–8% of castration-resistant tumors overall, an enrichment of approximately twofold over the frequency in localized prostate cancers (3–5). This enrichment suggests an association of ATM mutations with aggressive disease. Indeed, germline ATM mutations are significantly more common among patients with lethal compared with indolent prostate cancer (6) and more common among patients with high-grade disease (7), although whether this association persists for somatic ATM alterations is unknown.

Beyond acting as a potential prognostic biomarker in prostate cancer, ATM status may also be predictive of response to novel targeted therapies. Initial trials of the PARP inhibitor, olaparib, in metastatic castration-resistant prostate cancer (mCRPC) showed an impressive response rate among patients with mutations in the homologous recombination repair pathway, apparently including patients with ATM deficiency (8, 9). However, retrospective series and recent findings from prospective phase II and phase III trials examining a variety of PARP inhibitors (olaparib, rucaparib, niraparib, and talazoparib) in patients with mCRPC have shown that tumor responses, as well as progression-free survival estimates, are relatively modest in patients with the ATM mutation compared with patients with the BRCA1/2 mutation who appear to derive the majority of the benefit (10–14). This suggests that PARP inhibitor treatment may not induce synthetic lethality, or perhaps not to the same extent, in ATM-mutated prostate cancers compared with BRCA1/2-mutated cases (15, 16). However, the recent emergence of potent ATR (ataxia telangiectasia and Rad3-related protein) inhibitors has renewed interest in interrogating ATM status in prostate cancer and other malignancies (16–19), because initial phase I trials have shown favorable responses to ATR inhibitors that may be specific to ATM-deficient tumors (20). Preliminary studies also suggest a potential sensitivity of ATM-mutated prostate cancers to anti-PD-1 immunotherapy (21, 22), radium-223 treatment (23), and bipolar androgen therapy (24, 25). Therefore, the availability of an assay to detect ATM deficiency/loss would have great clinical utility in prioritizing such patients for clinical trials exploring these unique therapeutic vulnerabilities.

Before targeted therapies can be deployed for prostate tumors with ATM inactivation, numerous open questions about the clinical, pathologic, and genomic characteristics of ATM-deficient tumors will need to be resolved. For example, it remains unclear what fraction of patients with ATM germline or somatic mutations have detectable second-hit alterations and/or protein loss suggesting complete ATM inactivation and implying likely response to targeted therapy. In addition, how frequently are ATM alterations subclonal? What is the sensitivity of DNA-sequencing assays for detection of biallelic inactivation, and what assays should be used to screen patients for ATR inhibitor trial eligibility? Given that ATM inactivation and aberrations in other DNA damage repair pathway genes generally occur in the primary tumor rather than during metastatic evolution (26), many of these questions can be resolved by studying primary prostate cancers, which are more readily available compared with metastatic samples. Herein, we developed and genetically validated a clinical grade IHC assay to detect ATM protein loss and used it to assess ATM status in primary prostate cancers with known germline ATM mutations, as well as to screen for ATM inactivation in a cohort of more than 1,000 primary tumors. We comprehensively compared ATM protein status with DNA-sequencing data and characterized the largest group of ATM-deficient primary prostate cancers reported to date.

Patients and tissue samples

With Johns Hopkins Institutional Review Board (Baltimore, MD) approval, and in accordance with the U.S. Common Rule, three patient sets were included in this study. As this study involved only retrospective analysis of previously collected tissue samples, it was performed under a waiver of consent. (i) The first set included a cohort of all radical prostatectomies from 2004–2014 with primary Gleason pattern 5 and available clinical follow-up data (n = 77), of whom 52 had previously described targeted next-generation sequencing data from the UW OncoPlex platform available (27); these cases were used for genetic validation of the ATM IHC assay described below. (ii) The second patient set included 20 primary prostate tumors with available radical prostatectomy tissue with known pathogenic germline mutations in ATM. Of these cases, 16 had ATM mutations detected during sequencing of benign seminal vesicle or leukocyte DNA performed as a part of previously described studies (6), while the remaining four had ATM mutations detected using clinical grade germline sequencing platforms (Invitae, Color Genomics) from saliva samples (12). (iii) The third patient set included seven partially overlapping tissue microarray (TMA) cohorts from radical prostatectomies performed at Johns Hopkins (Baltimore, MD). These cases were used to screen for the frequency of ATM protein loss using the IHC assay. All of these TMA cohorts have been described previously, and were selected on the basis of risk factors. Some focused on high-risk features that enriched for adverse oncologic outcomes, so they do not represent an unbiased survey of the overall radical prostatectomy population at Johns Hopkins (Baltimore, MD). In brief, these consisted of: (i) a set of control patients who lacked a HOXB13 germline mutation, not selected by outcome (n = 99; ref. 28); (ii) a set of high-grade (Gleason score 9/10) tumors at radical prostatectomy from 1998 to 2005, designed for comparison with high-grade urothelial carcinomas, not selected by outcome (n = 38; ref. 29); (iii) a set of grade-matched African-American and European-American radical prostatectomy samples from 1995 to 2005, enriched for Gleason score ≥7 pathology, not selected by outcome (n = 292; refs. 30, 31); (iv) a set of grade-matched African-American and European-American radical prostatectomy samples from 2006 to 2010, all Gleason score 4+3 = 7 and higher, not selected by outcome (n = 163; refs. 30, 31); (v) a set of patients who all developed metastatic disease after radical prostatectomy at Johns Hopkins (Baltimore, MD) from 1995 to 2011, who were evaluated for response to abiraterone or enzalutamide (n = 34; ref. 32); (vi) a cohort study of men undergoing radical prostatectomy from 1992 to 2009 with intermediate- and high-risk disease who were followed for development of metastatic disease (n = 325; ref. 33); and (vii) a set of men with biochemical recurrence following radical prostatectomy from 1992 to 2009 (n = 240; ref. 34).

IHC

ATM protein IHC was performed using the Y170 rabbit monoclonal antibody (Abcam, 32420) on the Ventana Discovery XT Autostaining System (Roche/Ventana Medical Systems). The ATM Y170 clone was used to develop a similar IHC diagnostic assay (35) and was deployed in the phase III GOLD study, which looked at combination of olaparib and paclitaxel in patients with advanced gastric cancer who have progressed following first-line therapy (36). Slides were incubated with primary antibody (1:100 dilution) after antigen retrieval in CC1 buffer, and primary antibody incubation was followed by detection with the OptiView HQ System (Roche/Ventana Medical Systems). Each TMA spot or standard histologic section containing tumor cells was visually dichotomously scored for presence or absence of nuclear ATM signal by a urologic pathologist (T.L. Lotan). The pathologist was blinded to sequencing results when reading the IHC. A spot was considered to show ATM protein loss if any tumor cells in any tumor spot showed loss of nuclear ATM, with intact staining in admixed benign prostate glands and surrounding stromal cells, endothelial cells, or lymphocytes. Spots without internal control staining were considered ambiguous and were not scored. All samples were initially screened for ATM loss by scoring TMA spots; however, for all cases with ATM protein loss on TMA, confirmatory ATM immunostaining was also performed on standard histologic tissue sections.

ERG IHC was performed on the Ventana Platform (Ventana Discovery Ultra, Ventana Medical Systems) using a previously reported genetically validated staining and scoring protocol (37, 38). In brief, this assay utilized a rabbit anti-human ERG antibody (EPR3864). ERG was scored as positive if any tumor glands showed nuclear ERG expression, negative if no sampled tumor glands showed ERG expression, or ambiguous if endothelial nuclei did not show ERG expression within the evaluated tumor core as an internal positive control.

Cell lines

Isogenic CWR22Rv1 cell lines with and without biallelic ATM or BRCA2 inactivation were derived via CRISPR/Cas9 genome editing as described previously (39). Cells were pelleted and formalin-fixed, paraffin-embedded (FFPE) as described previously (40). Mycoplasma testing was conducted every 3 months (latest date October, 2019) and authentication of genome editing was described previously (39). Cells were passaged no more than twice before experiments were performed.

DNA isolation

On standard histologic sections, tumor tissue was macrodissected guided by hematoxylin and eosin (H&E)-stained section. Sections (5 × 10 μm) from FFPE tumor samples were used for DNA extraction. For the UW OncoPlex sequencing assay, DNA was extracted from FFPE material using the Qiagen FFPE DNA Extraction Kit according to the manufacturer's directions. DNA concentrations were quantified with the Qubit Fluorometer, using a Quant-iT dsDNA High Sensitivity Assay Kit (Invitrogen). For the Myriad HRD Plus sequencing assay, sections from FFPE tissue were first incubated in proteinase K followed by DNA extraction performed using the Promega Maxwell 16 LEV FFPE Plus Kit (AS1290, Promega) according to the manufacturer's instructions.

Targeted next-generation sequencing

Targeted next-generation sequencing was performed in a subset of cases using the UW OncoPlex assay as described previously (27), and for all cases we performed the HRD Plus Assay (Myriad Genetics). The HRD Plus assay has been described previously in detail for BRCA1/2 sequencing (41); here, it was modified to also include the entire coding region of ATM and a panel of 106 additional genes (Supplementary Table S1). Briefly, the HRD Plus assay uses a custom hybridization capture method employing Integrated DNA Technologies' xGen Hybridization Capture Technology (Integrated DNA Technologies). Sequencing was performed on an Illumina HiSeq2500 using a 200 cycle HiSeq Rapid SBS kit v2 and a HiSeq Rapid PE cluster kit v2.

Sequence analysis

Average coverage for ATM after alignment to the target regions and removal of nonclonal reads was 687 (range, 217–1,262). Novel variants identified by tumor sequencing using the HRD Plus assay were classified using a process that is consistent with the published standards and guidelines for clinical testing from the American College of Medical Genetics and Genomics (42). Variants were classified into one of five categories: deleterious, suspected deleterious, variant of uncertain significance, favor polymorphism, and polymorphism. Both deleterious and suspected deleterious variants were considered pathogenic. Variant classifications are stored in a classification database and can be retrieved each time they are observed during routine testing.

Statistical analysis

Clinicopathologic characteristics of tumors with and without ATM loss were compared using the χ2 test and the Wilcoxon rank-sum test for categorical and continuous variables, respectively. Metastasis-free survival (MFS) was estimated with Kaplan–Meier curves, both univariate and adjusted for Gleason score and TMA set (43). A proportional hazards frailty model was used to evaluate the prognostic effect of ATM loss adjusted for known prognostic factors, while accounting for clustering within TMA sets (44). All analyses were performed with SAS v9.4 (SAS Institute).

Genetic validation of ATM IHC assay

ATM IHC was initially validated using isogenic CWR22Rv1 cell lines with and without biallelic ATM inactivation derived via CRISPR/Cas9 genome editing (39). The clone with ATM knockout was previously confirmed to harbor a biallelic 33-nucleotide insertion in ATM (c.3383_3384ins33; p.Q1128_E1129insTASANSF*), resulting in a premature stop codon (39). These cells lacked detectable ATM protein by immunoblotting (Fig. 1A) and by IHC performed on a FFPE cell block with the same antibody clone (Fig. 1B). Next, we performed ATM IHC on a TMA containing 77 primary Gleason pattern 5 tumors procured from consecutive radical prostatectomies at our institution (27). Overall, 84% (65/77) were evaluable for ATM IHC, defined by adequate nuclear staining in internal control benign glands and stromal cells. Of these, 12% (8/65) had ATM protein loss. Of the 65 evaluable cases, 52 had previously generated UW OncoPlex sequencing data (27), including seven cases with ATM protein loss (13%; Fig. 1C). Of the seven cases with ATM protein loss and sequencing data available for comparison, four cases had a deleterious ATM mutation and two had shallow ATM genomic deletions (LOH implying monoallelic loss; Table 1; Fig. 1C). The final case had no detectable ATM alterations, although tumor content was noted to be low, precluding accurate copy-number calls. Of the four deleteriously mutated ATM cases, three had definitive LOH suggesting biallelic inactivation; the remaining case was uncertain due to low tumor content, however, biallelic loss was favored (Table 1). Of the remaining 45 cases without ATM protein loss, one had a pathogenic ATM missense mutation (p.R3008C) without LOH that was evident only on manual review of the sequencing data as the variant allele fraction was below the threshold of the variant calling algorithm. Of note, this mutation has been reported in clonal hematopoiesis of indeterminate potential and chronic lymphocytic leukemia and could represent a hematolymphoid clone given the low variant allele fraction (45). In summary, the negative predictive value of the ATM IHC assay for lack of underlying pathogenic ATM mutation was 98% (44/45) and the assay was 80% (4/5) sensitive for pathogenic ATM mutations and 100% (3/3) sensitive for biallelic ATM alteration.

Figure 1.

Genetic validation of ATM IHC assay. A, Immunoblotting for ATM in cell lysates of isogenic CWR22Rv1 cell lines with and without biallelic ATM or BRCA2 inactivation via CRISPR/Cas9 genome editing (40). B, ATM IHC in FFPE cell pellets of CWR22Rv1 cell lines with and without biallelic ATM inactivation. C, H&E staining (top row) and ATM IHC (bottom row) in representative primary Gleason pattern 5 prostate tumors with ATM protein loss and available ATM sequencing results (ref. 27; all images reduced from 200×). Benign glands (arrows) or stromal/endothelial nuclei provide an internal positive control in each case. KO, knockout.

Figure 1.

Genetic validation of ATM IHC assay. A, Immunoblotting for ATM in cell lysates of isogenic CWR22Rv1 cell lines with and without biallelic ATM or BRCA2 inactivation via CRISPR/Cas9 genome editing (40). B, ATM IHC in FFPE cell pellets of CWR22Rv1 cell lines with and without biallelic ATM inactivation. C, H&E staining (top row) and ATM IHC (bottom row) in representative primary Gleason pattern 5 prostate tumors with ATM protein loss and available ATM sequencing results (ref. 27; all images reduced from 200×). Benign glands (arrows) or stromal/endothelial nuclei provide an internal positive control in each case. KO, knockout.

Close modal
Table 1.

Next-generation sequencing results for tumors with ATM protein loss in primary Gleason pattern 5 cohort.

IDAlt ID (Ref 25)ATM IHCOncoPlex ATM mutationOncoPlex LOHOncoPlex germline suspected?Myriad ATM mutationMyriad LOHMyriad germline suspected
ATM26 61466 Loss LOH Yes No LOH Yes No 
ATM16 71497 Loss p.N845Ifs*Yes Yes p.N845Ifs*Yes No 
ATM19 72889 Loss p.K2589* Uncertain, biallelic favored* Yes p.K2589* Uncertain Uncertain 
ATM30 71513 Loss ∼28 bp indel that takes out the exon 11/intron 11 splice boundary Yes No c.1774_1802+4delins6 Yes No 
ATM31 71514 Loss p.H2195Qfs*Yes No p.H2195Qlnfs*No No 
ATM27 N/A Loss LOH Yes No LOH Yes No 
ATM42 71515 Heterogeneous loss None Uncertain* No LOH Yes No 
N/A 69210 Intact p.R3008C No No N/A N/A N/A 
IDAlt ID (Ref 25)ATM IHCOncoPlex ATM mutationOncoPlex LOHOncoPlex germline suspected?Myriad ATM mutationMyriad LOHMyriad germline suspected
ATM26 61466 Loss LOH Yes No LOH Yes No 
ATM16 71497 Loss p.N845Ifs*Yes Yes p.N845Ifs*Yes No 
ATM19 72889 Loss p.K2589* Uncertain, biallelic favored* Yes p.K2589* Uncertain Uncertain 
ATM30 71513 Loss ∼28 bp indel that takes out the exon 11/intron 11 splice boundary Yes No c.1774_1802+4delins6 Yes No 
ATM31 71514 Loss p.H2195Qfs*Yes No p.H2195Qlnfs*No No 
ATM27 N/A Loss LOH Yes No LOH Yes No 
ATM42 71515 Heterogeneous loss None Uncertain* No LOH Yes No 
N/A 69210 Intact p.R3008C No No N/A N/A N/A 

Abbreviation: N/A, not assessed.

*Low tumor content.

To verify these sequencing results on a different platform, we reisolated DNA and resequenced all seven samples with ATM protein loss, this time using the Myriad HRD Plus assay. All four pathogenic mutations were confirmed (of which two had definitive LOH; Table 1). The additional two samples with isolated LOH by the OncoPlex assay also showed isolated LOH by the Myriad assay and the single sample that was previously low tumor content and without detectable alterations on the OncoPlex assay showed ATM LOH on Myriad resequencing (Table 1). Taking the results from both DNA-sequencing assays together, all seven cases with ATM protein loss had evidence of at least monoallelic ATM inactivation, however, only three of these cases had evidence of biallelic inactivation of ATM by one or both sequencing assays. These data suggest that targeted next-generation sequencing assays may be insensitive for biallelic deleterious ATM genomic (or epigenomic) alterations leading to protein loss and may be complemented by IHC staining assays.

ATM protein expression and somatic ATM status in primary prostate tumors with germline pathogenic ATM mutations

Next, we assessed the prevalence of ATM protein loss in primary prostate cancer cases with known ATM germline mutations. We identified 20 cases with pathogenic germline ATM mutations and available primary tumor tissue at our institution (6, 12). Tumor tissue for all cases was resequenced on the Myriad HRD Plus platform and mutations were confirmed in 19 of 20 cases (Table 2). These cases were generally high-grade, high-stage tumors, with more than half harboring Gleason grade group 5 cancer (Table 2). Of the 19 cases with confirmed germline ATM alterations, 74% (14/19) had ATM protein loss upon immunostaining of standard histologic sections of the dominant tumor nodule. Of the cases with ATM protein loss, 79% (11/14) had homogeneous loss in the entire sampled dominant tumor nodule, suggesting that the ATM mutation was likely an early clonal event (Fig. 2). Among the cases with ATM protein loss, which were evaluable for copy-number alteration or a second pathogenic ATM alteration, 70% (7/10) had genomic evidence of potential biallelic inactivation. The term “potential” biallelic inactivation is used because in cases with two ATM alterations, the phase (cis vs. trans) of the mutations was not conclusively determined (Table 2). Of interest, two of the three cases with protein loss, but lacking evidence of LOH, had a second ATM mutation detected that was classified as a variant of unknown significance (VUS; p.W3055C; p.A1272N), suggesting the potential for biallelic ATM inactivation in these cases as well. In contrast, among five cases with germline ATM mutations and intact ATM protein, four were evaluable for copy-number alteration and none had an evidence of LOH or a second somatic ATM mutation, suggesting the development of a sporadic prostate cancer in individuals with an incidental ATM germline lesion.

Table 2.

Genomic and clinicopathologic characteristics of prostate tumors with pathogenic germline ATM mutations.

IDATM IHCATM mutation from germline sequencingGermline sequencing platformATM mutation from tumor sequencingMyriad LOHTwo pathogenic alterations in ATM?AgeRaceGleason (primary)Gleason (secondary)Gleason (sum)Pathologic stage
ATM6 Loss p.L2005fs* Research p.Q2007Rfs*11 + p.W3055C (VUS) No No 74 T3a N1 MX 
ATM11 Heterogeneous loss p.K750K Research p.K750K + p.A1272N (VUS) No No 63 T2 N0 MX 
ATM13 Loss S1905I fs*25 Clinical p.S1905Ifs*25 No No 44 T3b N0 MX 
ATM7 Loss p.P292L Research p.P292L + p.C430* No Yes 66 T3a N0 MX 
ATM3 Loss p.D2795fs* Research p.F2799Kfs*4 Yes Yes 64 T2 N0 MX 
ATM5 Heterogeneous loss c.8988–1G>A Research c.8988–1G>A Yes Yes 53 T3a N0 MX 
ATM10 Loss p.K750K Research p.K750K Yes Yes 64 T2 N0 MX 
ATM12 Loss Duplication exon 48 Clinical Large genomic rearrangement + p.I2356Vfs*2 Uncertain Yes 55 T3b N0 MX 
ATM15 Loss p.K2756* Research p.K2756* + del exons 33-62 No Yes 61 T3b N1 MX 
ATM20 Loss p.L1107* Research p.L1107* Yes Yes 60 T2 N0 MX 
ATM18 Loss p.K2756* Research p.K2756* Uncertain Uncertain 54 T3a N0 MX 
ATM8 Loss p.G1458fs* Research p.G1458Qfs*15 Uncertain Uncertain 75 T3a N0 MX 
ATM1 Heterogeneous loss p.T452fs* Research p.T452Nfs*21 Uncertain Uncertain 52 T2 N0 MX 
ATM14 Loss c.8786+1G>A Research c.8786+1G>A Uncertain Uncertain 50 T3a N0 MX 
ATM4 Intact p.R2598* Research p.R2598* No No 69 T3a N0 MX 
ATM9 Intact p.2546_2548del Research p.R2547_S2549del No No 49 T3a N0 MX 
ATM17 Intact p.S274fs* Research p.L275* No No 47 T2 N0 MX 
ATM21 Intact c.7629 +2 T>C Clinical c.7629+2T>C Uncertain Uncertain 60 T3a N0 MX 
ATM22 Intact p.S160Afs*23 Clinical p.S160Afs*23 No No 65 T3b N1 MX 
ATM2 Intact p.R447* Research No variants found Uncertain Uncertain 36 T2 N0 MX 
IDATM IHCATM mutation from germline sequencingGermline sequencing platformATM mutation from tumor sequencingMyriad LOHTwo pathogenic alterations in ATM?AgeRaceGleason (primary)Gleason (secondary)Gleason (sum)Pathologic stage
ATM6 Loss p.L2005fs* Research p.Q2007Rfs*11 + p.W3055C (VUS) No No 74 T3a N1 MX 
ATM11 Heterogeneous loss p.K750K Research p.K750K + p.A1272N (VUS) No No 63 T2 N0 MX 
ATM13 Loss S1905I fs*25 Clinical p.S1905Ifs*25 No No 44 T3b N0 MX 
ATM7 Loss p.P292L Research p.P292L + p.C430* No Yes 66 T3a N0 MX 
ATM3 Loss p.D2795fs* Research p.F2799Kfs*4 Yes Yes 64 T2 N0 MX 
ATM5 Heterogeneous loss c.8988–1G>A Research c.8988–1G>A Yes Yes 53 T3a N0 MX 
ATM10 Loss p.K750K Research p.K750K Yes Yes 64 T2 N0 MX 
ATM12 Loss Duplication exon 48 Clinical Large genomic rearrangement + p.I2356Vfs*2 Uncertain Yes 55 T3b N0 MX 
ATM15 Loss p.K2756* Research p.K2756* + del exons 33-62 No Yes 61 T3b N1 MX 
ATM20 Loss p.L1107* Research p.L1107* Yes Yes 60 T2 N0 MX 
ATM18 Loss p.K2756* Research p.K2756* Uncertain Uncertain 54 T3a N0 MX 
ATM8 Loss p.G1458fs* Research p.G1458Qfs*15 Uncertain Uncertain 75 T3a N0 MX 
ATM1 Heterogeneous loss p.T452fs* Research p.T452Nfs*21 Uncertain Uncertain 52 T2 N0 MX 
ATM14 Loss c.8786+1G>A Research c.8786+1G>A Uncertain Uncertain 50 T3a N0 MX 
ATM4 Intact p.R2598* Research p.R2598* No No 69 T3a N0 MX 
ATM9 Intact p.2546_2548del Research p.R2547_S2549del No No 49 T3a N0 MX 
ATM17 Intact p.S274fs* Research p.L275* No No 47 T2 N0 MX 
ATM21 Intact c.7629 +2 T>C Clinical c.7629+2T>C Uncertain Uncertain 60 T3a N0 MX 
ATM22 Intact p.S160Afs*23 Clinical p.S160Afs*23 No No 65 T3b N1 MX 
ATM2 Intact p.R447* Research No variants found Uncertain Uncertain 36 T2 N0 MX 

Abbreviation: W, white.

Figure 2.

Primary prostate tumors with heterogeneous ATM protein loss. Heterogeneous ATM immunostaining (left column) in representative tumors with germline or inferred somatic ATM mutation. ERG immunoreactivity (right column), indicating underlying ERG gene rearrangement, is homogeneous in all cases, suggesting the ERG rearrangement preceded subclonal ATM inactivation (all images reduced from 200×).

Figure 2.

Primary prostate tumors with heterogeneous ATM protein loss. Heterogeneous ATM immunostaining (left column) in representative tumors with germline or inferred somatic ATM mutation. ERG immunoreactivity (right column), indicating underlying ERG gene rearrangement, is homogeneous in all cases, suggesting the ERG rearrangement preceded subclonal ATM inactivation (all images reduced from 200×).

Close modal

Prevalence of ATM loss among large set of surgically treated patients

Next, we assessed the prevalence of ATM protein loss in several previously described sets of TMAs prepared from genetically unselected radical prostatectomy specimens. There were a total of 1,076 patients with tumor tissue sampled in these TMAs, which included the primary Gleason pattern 5 TMA set described above. Overall, 82% (885/1,076) of these samples were interpretable for ATM staining and the subset of 831 with complete clinicopathologic data and clinical follow-up information are presented in Table 3. ATM loss was present in 3% (25/831) of these evaluable tumors overall. Patients with ATM loss underwent prostatectomy more recently (P = 0.028), had higher Gleason grade (P < 0.0001), pathologic stage (P = 0.024), and CAPRA-S score (P = 0.002). ATM loss was identified more commonly in Grade group 5 (17/181; 9%) compared with all other grades (8/650; 1%; P < 0.0001). Of the cases with ATM loss, seven were from the primary Gleason pattern 5 TMA and one case overlapped with the germline cases described above, and were already sequenced (Tables 1 and 2). The remaining 17 cases were processed for DNA sequencing and restaining for ATM using standard histologic sections to assess for ATM protein expression heterogeneity (Table 4; Fig. 2). Of these, 80% (4/5) with homogeneous ATM protein loss and 50% (6/12) with heterogeneous ATM protein loss had detectable pathogenic ATM alterations. One additional case with heterogeneous protein loss had a mutation potentially affecting a canonical splice site in ATM (c.6808-1G>C), suggesting pathogenicity (Fig. 2). Of the mutated cases, only one was inferred to have a germline pathogenic ATM mutation based on variant allele frequency in the tumor sequencing, while the remainder were inferred somatic alterations or indeterminate between germline and somatic based on tumor sequencing. Among the mutated cases with homogeneous protein loss and evaluable copy-number alteration data, two of two had potential biallelic inactivation of ATM. Among the mutated cases with heterogeneous protein loss and evaluable copy-number alteration data, three of five had potential biallelic inactivation, with one additional case harboring an additional VUS in ATM.

Table 3.

Clinicopathologic features of cases with and without ATM loss on TMA screening.

VariableATM loss (n = 25)aATM intact (n = 806)aP
Age (years), median (IQR) 63 (56–66) 59 (55–64) 0.073 
Race, n (%) 
 White 23 (92%) 599 (74%) 0.143 
 Black 2 (8%) 193 (24%)  
 Other 14 (2%)  
RP year, median (IQR) 2004 (2001–2008) 2001 (1998–2005) 0.028 
PSA (ng/mL), median (IQR) 7.7 (5.9–12.8) 7.3 (5.0–11.5) 0.271 
RP grade group, n (%) 
 1 (Gleason score 6) 117 (15%) <0.0001 
 2 (Gleason score 3+4) 3 (12%) 209 (26%)  
 3 (Gleason score 4+3) 2 (8%) 214 (27%)  
 4 (Gleason score 8) 3 (12%) 102 (13%)  
 5 (Gleason score 9–10) 17 (68%) 164 (20%)  
Pathologic stage, n (%) 
 T2 N0 5 (20%) 281 (35%) 0.024 
 T3a N0 9 (36%) 310 (39%)  
 T3b N0 9 (36%) 106 (13%)  
 Tx N1 2 (8%) 101 (13%)  
CAPRA-S 6.0 (4.0–9.0) 4.0 (3.0–6.0) 0.002 
VariableATM loss (n = 25)aATM intact (n = 806)aP
Age (years), median (IQR) 63 (56–66) 59 (55–64) 0.073 
Race, n (%) 
 White 23 (92%) 599 (74%) 0.143 
 Black 2 (8%) 193 (24%)  
 Other 14 (2%)  
RP year, median (IQR) 2004 (2001–2008) 2001 (1998–2005) 0.028 
PSA (ng/mL), median (IQR) 7.7 (5.9–12.8) 7.3 (5.0–11.5) 0.271 
RP grade group, n (%) 
 1 (Gleason score 6) 117 (15%) <0.0001 
 2 (Gleason score 3+4) 3 (12%) 209 (26%)  
 3 (Gleason score 4+3) 2 (8%) 214 (27%)  
 4 (Gleason score 8) 3 (12%) 102 (13%)  
 5 (Gleason score 9–10) 17 (68%) 164 (20%)  
Pathologic stage, n (%) 
 T2 N0 5 (20%) 281 (35%) 0.024 
 T3a N0 9 (36%) 310 (39%)  
 T3b N0 9 (36%) 106 (13%)  
 Tx N1 2 (8%) 101 (13%)  
CAPRA-S 6.0 (4.0–9.0) 4.0 (3.0–6.0) 0.002 

Abbreviations: IQR, interquartile range; RP, prostatectomy.

aSample sizes vary due to missing values.

Table 4.

Genomic and clinicopathologic features of tumors with ATM protein loss on TMA screening.

IDATM IHCATM mutation from tumor sequencingGermline or somatic (inferred)LOHTwo pathogenic alterations in ATM?AgeRaceGleason (primary)Gleason (secondary)Gleason (sum)Pathologic stage
ATM25 Loss p.L1722Rfs*25 + c.2124+1G>A Somatic No Yes 71 T3b N0 MX 
ATM23 Loss p.Q1361* Somatic Uncertain Uncertain 57 T3b N0 MX 
ATM28 Loss p.R447* + del exons 1-32 Germline and somatic No Yes 65 T3b N1 MX 
ATM24 Loss p.S743* Somatic Uncertain Uncertain 59 T3b N0 MX 
ATM29 Loss None None  No 66 T3a N0 MX 
ATM38 Heterogeneous loss c.185+1G>A Somatic Yes Yes 56 T3b N0 MX 
ATM41 Heterogeneous loss c.6808-1G>C Uncertain Uncertain Uncertain 63 T2 N0 MX 
ATM40 Heterogeneous loss Complex large rearrangement + p.V2766K (VUS) Somatic No No 55 T3a N0 MX 
ATM37 Heterogeneous loss p.Q2220Rfs*15 Somatic No No 59 T2 N0 MX 
ATM33 Heterogeneous loss p.Q513* Uncertain Uncertain Uncertain 58 T2 N0 MX 
ATM39 Heterogeneous loss p.Q893* Somatic Yes Yes 67 T3b N0 MX 
ATM32 Heterogeneous loss Large genomic rearrangement Somatic Yes Yes 67 T3a N0 MX 
ATM34 Heterogeneous loss None None  No 72 T2 N0 MX 
ATM35 Heterogeneous loss None None  No 70 T3a N0 MX 
ATM36 Heterogeneous loss None None  No 51 T2 N0 MX 
ATM43 Heterogeneous loss None None  No 52 T3b N0 MX 
ATM44 Heterogeneous loss None None  No 49 T3b N1 MX 
IDATM IHCATM mutation from tumor sequencingGermline or somatic (inferred)LOHTwo pathogenic alterations in ATM?AgeRaceGleason (primary)Gleason (secondary)Gleason (sum)Pathologic stage
ATM25 Loss p.L1722Rfs*25 + c.2124+1G>A Somatic No Yes 71 T3b N0 MX 
ATM23 Loss p.Q1361* Somatic Uncertain Uncertain 57 T3b N0 MX 
ATM28 Loss p.R447* + del exons 1-32 Germline and somatic No Yes 65 T3b N1 MX 
ATM24 Loss p.S743* Somatic Uncertain Uncertain 59 T3b N0 MX 
ATM29 Loss None None  No 66 T3a N0 MX 
ATM38 Heterogeneous loss c.185+1G>A Somatic Yes Yes 56 T3b N0 MX 
ATM41 Heterogeneous loss c.6808-1G>C Uncertain Uncertain Uncertain 63 T2 N0 MX 
ATM40 Heterogeneous loss Complex large rearrangement + p.V2766K (VUS) Somatic No No 55 T3a N0 MX 
ATM37 Heterogeneous loss p.Q2220Rfs*15 Somatic No No 59 T2 N0 MX 
ATM33 Heterogeneous loss p.Q513* Uncertain Uncertain Uncertain 58 T2 N0 MX 
ATM39 Heterogeneous loss p.Q893* Somatic Yes Yes 67 T3b N0 MX 
ATM32 Heterogeneous loss Large genomic rearrangement Somatic Yes Yes 67 T3a N0 MX 
ATM34 Heterogeneous loss None None  No 72 T2 N0 MX 
ATM35 Heterogeneous loss None None  No 70 T3a N0 MX 
ATM36 Heterogeneous loss None None  No 51 T2 N0 MX 
ATM43 Heterogeneous loss None None  No 52 T3b N0 MX 
ATM44 Heterogeneous loss None None  No 49 T3b N1 MX 

Abbreviation: W, white.

Clinical outcomes of primary prostate tumors with ATM protein loss

Because many of the TMA sets that we stained for ATM had clinical follow-up information available after radical prostatectomy, we compared biochemical recurrence-free survival (BCR-FS), MFS, and prostate cancer–specific survival (PCSS) in men with and without ATM protein loss from the TMA sets. As described above, the prevalence of ATM loss was strongly correlated with Gleason grade group. In addition, the different TMA sets varied considerably both in the proportion of cases with ATM loss and with respect to survival probabilities based on their varied design and differing study populations. Thus, while ATM loss was significantly associated with an increased risk of metastasis in univariate analysis [HR, 2.32; 95% confidence interval (CI), 1.23–4.39; P = 0.009], it was not significantly associated with increased risk of metastasis after adjusting for Gleason grade and including TMA set as a random variable in a proportional hazards frailty model (adjusted HR, 1.06; 95% CI, 0.55–2.05; P = 0.857). Similar results were seen with a standard (unadjusted) and adjusted Kaplan–Meier analysis (Supplementary Fig. S1). Furthermore, ATM loss was not significantly associated with BCR-FS, MFS, or PCSS in random-effects Cox models adjusted for age, race (White vs. non-White), radical prostatectomy year, and Gleason grade group (Supplementary Table S2). Concordant results were observed if CAPRA-S replaced Gleason grade group (data not shown).

Timing of ATM loss in primary prostate cancer progression

The higher frequency of ATM loss heterogeneity among cases in the TMA study (71% or 12/17) compared with those in the germline study (21% or 3/14) was interesting and suggested that ATM inactivation may occur later in tumors with inferred somatic mutations (which comprised the majority in the TMA study) compared with those with germline mutations. However, given that multifocal and multiclonal tumors are common in primary prostate cancer, it is also conceivable that the tumors with heterogeneous ATM expression represent collisions between independent clones. To resolve this, and to begin to elucidate the timing of ATM loss in primary prostate cancer, we assessed ERG status in cases with heterogeneous ATM loss. ERG gene rearrangements are present in approximately half of all prostate cancer cases in European ancestry populations (46) and are likely among the earliest genomic alterations during prostatic tumorigenesis, occurring in some cases of prostatic intraepithelial neoplasia (PIN; refs. 47–49) and before PTEN genomic deletion (50, 51). Accordingly, ERG expression (a highly validated surrogate marker of underlying ERG gene rearrangement; ref. 38) is almost always ubiquitously present in all tumor cells from a given tumor nodule if ERG is rearranged, unless the tumor represents a collision between two independent clones (51). Among cases with heterogeneous ATM loss (including three germline cases and 12 from the TMA study described above), ERG was expressed in 53% (8/15) of cases and was uniformly expressed in both ATM-positive and ATM-negative tumor cells in all cases, including two germline cases and six TMA cases (Fig. 2). These data strongly suggest that in cases with inferred somatic ATM genomic alteration (the majority in the TMA study), ATM inactivation commonly occurs subclonally and subsequent to ERG gene rearrangement, and that tumors with heterogeneous ATM expression do not commonly represent collisions of independent clones.

Because ATM loss was most commonly homogeneous and apparently clonal in primary prostate tumors with pathogenic germline ATM mutations, this raised the question of how early ATM inactivation may occur in patients with germline mutations. To begin to assess this, we screened 16 ATM protein-negative cases with known or inferred germline ATM alterations from the above cohorts for high-grade PIN, the presumptive precursor for invasive carcinoma in the prostate. We exclusively selected PIN lesions present on slides without invasive tumor by H&E, because recent studies have suggested that retrograde intraductal spread of invasive carcinoma can masquerade as PIN (52). We identified 11 cases that met these criteria, of which nine (82%) were evaluable for ATM IHC in the PIN lesions. Only one of nine (11%) showed ATM loss in the PIN (Fig. 3). Taken together, with our finding that ATM loss in germline cases is most commonly homogeneous and likely clonal, these findings suggest that ATM loss commonly occurs at or just after initial tumor invasion, rather than in precursor lesions in the prostate.

Figure 3.

ATM expression in high-grade PIN. A, Focus of PIN from prostatectomy showing intact ATM. This prostatectomy specimen harbored invasive tumor in additional tissue blocks with homogeneous ATM loss and two pathogenic ATM mutations [ATM p. P292L (germline) and p.C430*; all images reduced from 200×). B, Focus of PIN showing ATM loss in luminal cells, with expression of ATM in surrounding basal cells (arrowheads, top). Inset shows multiplex immunostaining for PIN4 (p63 and high molecular weight cytokeratin in brown highlight basal cells, while racemase in red highlights luminal PIN cells). There are two nearby atypical glands with a patchy basal cell layer on PIN4 IHC also showing loss of ATM (arrows). This prostatectomy specimen harbored invasive tumor in additional tissue blocks with homogeneous ATM loss and a pathogenic ATM germline mutation (ATM p.G1458Qfs*15; all images reduced from 200×). Higher magnification view of adjacent area to that depicted in top panels shows ATM loss in focal area of PIN with enlarged nucleoli (arrowheads, bottom). Inset shows multiplex immunostaining for PIN4 (p63 and high molecular weight cytokeratin in brown highlight basal cells, while racemase in red highlights luminal PIN cells). ATM is intact in surrounding, morphologically benign-appearing cells (arrows; all images reduced from 630×).

Figure 3.

ATM expression in high-grade PIN. A, Focus of PIN from prostatectomy showing intact ATM. This prostatectomy specimen harbored invasive tumor in additional tissue blocks with homogeneous ATM loss and two pathogenic ATM mutations [ATM p. P292L (germline) and p.C430*; all images reduced from 200×). B, Focus of PIN showing ATM loss in luminal cells, with expression of ATM in surrounding basal cells (arrowheads, top). Inset shows multiplex immunostaining for PIN4 (p63 and high molecular weight cytokeratin in brown highlight basal cells, while racemase in red highlights luminal PIN cells). There are two nearby atypical glands with a patchy basal cell layer on PIN4 IHC also showing loss of ATM (arrows). This prostatectomy specimen harbored invasive tumor in additional tissue blocks with homogeneous ATM loss and a pathogenic ATM germline mutation (ATM p.G1458Qfs*15; all images reduced from 200×). Higher magnification view of adjacent area to that depicted in top panels shows ATM loss in focal area of PIN with enlarged nucleoli (arrowheads, bottom). Inset shows multiplex immunostaining for PIN4 (p63 and high molecular weight cytokeratin in brown highlight basal cells, while racemase in red highlights luminal PIN cells). ATM is intact in surrounding, morphologically benign-appearing cells (arrows; all images reduced from 630×).

Close modal

Pathogenic mutations in the homologous recombination DNA repair pathway, including the BRCA2, BRCA1, and ATM genes, are common in advanced prostate cancer, occurring in nearly 20% of mCRPC cases (4). Nearly half of these alterations have proven to be inherited at the germline level, comprising close to 10% of men with mCRPC (3, 4). Historically, the BRCA2, BRCA1, and ATM genes have been grouped together in most analyses, given that all encode proteins that are key components of homology-mediated DNA repair. The first large-scale sequencing studies in prostate cancer showed that there is a twofold enrichment of mutations in these genes in metastatic compared with primary cancers (4, 53), suggesting that these alterations are associated with aggressive disease. Indeed, a number of earlier studies had already confirmed this hypothesis for germline BRCA2 mutations (54–57). More recent studies have found that germline alterations in BRCA2 and ATM (considered together) are significantly more common in lethal compared with indolent primary prostate cancer (6), are associated with grade reclassification in active surveillance cohorts (58), and with high-grade disease in surgical cohorts (7). The correlation of germline BRCA2 and ATM mutations with adverse pathologic features, including more advanced Gleason scores and higher PSA levels (6), likely drives a large part of this association with adverse outcomes. Considering all homologous repair gene mutations in aggregate, our group has previously shown that aggressive histologic subsets of primary prostate cancer, such as ductal carcinomas (59), primary Gleason pattern 5 disease (27), and intraductal cancer (60) have mutation rates approaching or exceeding those in metastatic disease.

While nearly all prior studies have considered BRCA2 and ATM mutations together because both genes encode proteins functioning in the homologous DNA repair pathway, emerging evidence from clinical trials suggests that the roles of these two genes may in fact be quite distinctive. Prospective phase II and phase III trials of PARP inhibitors (including olaparib, rucaparib, niraparib, and talazoparib) in patients with CRPC have shown that the response rates are much more limited in patients with the ATM mutation compared with patients with the BRCA2 mutation (9–13, 61). ATM is a sensor of double-strand DNA damage, and is a kinase with a diverse range of substrates; these clinical data suggest that some of its key functions in prostate cancer may be independent of its role in homology-mediated DNA repair. In fact, ATM has also been shown to modulate growth factor signaling and to play a role in other stress response pathways (2). Accordingly, our study is the first to examine the clinicopathologic features and outcomes of ATM-deficient primary prostate tumors separately from those with BRCA2 deficiency. This work was largely made possible by the development of a robust clinical grade ATM IHC assay, which we genetically validated and subsequently used to screen hundreds of prostate tumors for ATM loss. Importantly, we confirm that ATM loss is highly enriched in Grade group 5 prostate cancers and is associated with poor outcomes in univariate analyses of surgically treated patients. However, this association with aggressive disease appears to be largely driven by a correlation with high-grade group, and we show here that ATM loss is not independently prognostic of outcomes in multivariate models, although these data require confirmation in validation cohorts.

Although ATM loss appears not to be prognostic in primary prostate cancer, it will still be critically important to identify ATM-deficient prostate tumors for trials of targeted therapies, including ATR inhibitors as well as other rational therapies (e.g., radium-223). Similar to ATM, ATR is a PI3K-like kinase, however, unlike ATM, which responds largely to double-strand DNA breaks, ATR responds to single-strand breaks and other types of DNA damage. Because of its atypical nature, ATR inhibitors have historically lagged in development behind other serine–threonine kinase inhibitors, such as AKT inhibitors (18). However, several potent and relatively specific ATR inhibitors have been characterized and have shown synthetic lethal activity in ATM-deficient tumors in vitro and in early clinical trials (18, 19). Given these promising initial results, larger biomarker-selected trials are currently ongoing (e.g., NCT04095273) and will require screening of large populations of patients with prostate cancer for inclusion. Combined with previous germline cohorts (7), this study demonstrates that screening of grade group 5 tumors will likely be most fruitful for trial recruitment, with 9% of such patients (and as many as 13% of primary Gleason pattern 5 tumors) harboring ATM loss.

Equally important to defining which populations to screen for these trials is determining which assay will be utilized for the screening protocol. Our data strongly suggest that if germline ATM sequencing is used for enrollment, at least a quarter of enrolled patients with prostate cancer may be unlikely to respond because they lack ATM protein loss and biallelic inactivation and most likely have sporadic cancers. We found that the presence of ATM protein loss strongly enriched for underlying potential biallelic inactivation of ATM, with 70% of cases with ATM protein loss (and none of the cases without protein loss) harboring a likely somatic “second hit” in the ATM gene by DNA sequencing. It will require careful clinical trials to resolve whether ATM may function in a haploinsufficient manner as seen with other tumor suppressor genes (62) and to discern whether patients with monoallelic inactivation may also respond to targeted therapy similarly to those with biallelic inactivation. In this setting, ATM IHC will likely prove very useful to determine whether ATM deficiency is likely to be monoallelic or biallelic, because next-generation sequencing may miss complex rearrangements of the gene or epigenomic gene silencing and germline sequencing will not detect somatic or epigenomic ATM inactivation. If ATM is not a haploinsufficient tumor suppressor gene, then screening with germline DNA assays alone will be inadequate for enrollment in biomarker-selected clinical trials and the addition of somatic sequencing with ATM IHC assays will also be important. While there have been limited preclinical studies examining ATM happloinsufficiency in prostate cancer, previously published studies in breast cancer mouse models seem to suggest that Atm can function in a haploinsufficient manner in some genetic contexts (63, 64). In contrast, other preclinical studies found no increased mutagenic response to radiation in mice with hemizygous loss of Atm in germline and somatic tissue (65, 66), implying potential context-dependent effects. Additional work in prostate cancer preclinical models will be important to evaluate for potential evidence of ATM haploinsufficiency.

Our study is among the first to shed light on the underlying heterogeneity in ATM-deficient tumors and the likely timing of ATM loss in the setting of germline and somatic pathogenic mutations. Among tumors with germline ATM mutations, only 21% had heterogeneous ATM loss in the dominant nodule of the primary tumor, suggesting subclonal inactivation. Thus, when ATM loss occurs in patients with germline ATM mutations, it is most commonly a relatively early driver event, similar to ERG gene rearrangement. Analogous to ERG rearrangement, which is only infrequently seen in PIN (49), we demonstrate that ATM is only rarely lost in the presumptive prostate cancer precursor lesion (PIN) in cases with germline and apparent clonal ATM inactivation. This suggests that similar to ERG rearrangement, ATM loss likely occurs just after the point of tumor invasion. In contrast to these germline cases, in the TMA study where most ATM alterations were inferred to be somatic (based on variant allele frequency), nearly 70% of cases had heterogeneous or likely subclonal ATM loss. In cases with heterogeneous ATM loss, we demonstrate that ATM inactivation most likely occurred after ERG gene rearrangement given that ERG was expressed in all tumor cells.

Whether the presence of subclonal loss implies that patients are less likely to respond to targeted therapy is an unanswered question that will hopefully be resolved in future trials of ATR inhibitors and other molecularly targeted approaches. Studies to date have suggested that DNA repair gene mutations are most commonly present in the primary tumor and all metastases (26, 67), implying that they are relatively early drivers in most cases. If this is the case, then even patients with heterogeneous ATM loss in the primary tumor may have metastases with homogeneous ATM loss that will respond to therapy. On the other hand, it is conceivable that some cases may have late subclonal loss in the primary tumor, potentially even after metastatic spread has occurred, in which case response to targeted therapy may not be assured and testing of the metastatic deposit will be critical. In an attempt to shed light on these possibilities, we examined the medical records of all cases in this study with heterogeneous ATM protein loss in the primary tumor to identify cases with available metastatic tissue samples. Unfortunately, no cases had available tissue from metachronous metastases, although one case had tissue sampled from a synchronous pelvic lymph node metastasis (ATM44). Interestingly, in this case, there was clear heterogeneous ATM loss in the tumor, with intact ATM in a synchronous pelvic lymph node metastasis (Supplementary Fig. S2). Notably, this case did not have a detectable ATM mutation in the primary tumor, likely due to the subclonal nature of ATM loss identified by IHC. Consistent with this, cases that had heterogeneous ATM loss in our study were less likely to have any detectable underlying ATM mutation (50%), compared with those with homogeneous ATM protein loss (80%), highlighting the challenges of utilizing DNA-sequencing assays in potentially genetically heterogeneous tumors. This is particularly true in prostate cancer, where copy-number alterations and rearrangements are common causes of somatic genomic inactivation and may be missed by sequencing in a mixed population of tumor cells (68). Thus, while IHC may be useful to screen primary tumors for subclonal loss, it is equally important to follow-up in heterogeneous cases with sequencing or IHC evaluation of metastatic deposits to further evaluate trial eligibility.

Our study has some limitations that warrant discussion. First, all sequencing was done using a panel-based approach, where there may be limited sensitivity for shallow copy-number alterations and complex genomic rearrangements which can lead to ATM deficiency. This could be a possible explanation for cases with ATM IHC loss, but lacking an apparent genomic alteration. Although less likely, epigenomic silencing of the ATM locus remains another possibility. Second, although we screened a very large cohort of primary prostatectomies for ATM loss, this study was done using selected TMAs, which may not be representative of underlying population or tumor heterogeneity. The analyses of clinical outcomes are based on the combined data from nine TMA sets, which differ with respect to design, clinical characteristics, dates of surgery, postprostatectomy treatment, and time of TMA construction. Although the data are retrospective, patients were selected for each TMA on the basis of clinical characteristics rather than outcome, reducing differences in study design. Furthermore, incorporation of TMA set as a random effect in the proportional hazards frailty models addresses the clustering within TMA. Despite the large sample size, only 25 patients exhibited ATM loss, which may have affected stability of multivariate models. Importantly, the lack of a validation cohort in the prognostic analyses is a significant limitation and additional studies in independent cohorts are required to establish the validity of our findings.

In conclusion, we present the largest study to date using a genetically validated ATM protein IHC assay to interrogate more than 1,000 primary prostate cancers for ATM deficiency. We show that ATM loss is strongly associated with the highest Gleason grades, and that the negative prognostic impact of ATM loss is primarily driven by Gleason grade. Because not all ATM-mutated prostate cancers demonstrate ATM protein loss, and not all cases with protein loss have underlying genomic ATM alterations, we propose a combined interrogation approach utilizing both ATM IHC and next-generation sequencing assays for upcoming clinical trials. These trials will make possible a head-to-head comparison of the assays to select the most predictive biomarker. We anticipate that some combination of these assays will lead to optimal detection of ATM inactivation in prostate cancer for consideration of genomically targeted strategies in this subset of patients.

D.C. Salles reports non-financial support from Myriad Genetics during the conduct of the study. C.C. Pritchard reports personal fees from AstraZeneca outside the submitted work. A.M. De Marzo reports grants from NIH, NCI and The Department of Defense during the conduct of the study, Janssen Research and Development (sponsored research project unrelated to this manuscript) and Myriad Genetics (sponsored research project unrelated to this manuscript), and personal fees from Cepheid Inc (consulting fees, for project unrelated to this manuscript) outside the submitted work. J.S. Lanchbury reports personal fees from Myriad Genetics, Inc. (employee and stockholder) during the conduct of the study. K.M. Timms reports personal fees from Myriad Genetics, Inc. (employee and stockholder) during the conduct of the study and outside the submitted work. E.S. Antonarakis reports grants and personal fees from Janssen, and Sanofi, Dendreon, Merck, Bristol Myers Squibb, and AstraZeneca outside the submitted work, personal fees from Pfizer and Clovis, Eli Lilly, and Amgen outside the submitted work, grants from Johnson & Johnson, Genentech, Novartis, and Constellation outside the submitted work, and has a patent for AR-V7 liquid biopsy technology issued, licensed, and with royalties paid from Qiagen. T.L. Lotan reports non-financial support from Myriad Genetics (provided sequencing assays used) during the conduct of the study and grants from Roche/Ventana outside the submitted work. No potential conflicts of interest were disclosed by the other authors.

H. Kaur: Conceptualization, data curation, investigation, writing-original draft, writing-review and editing. D.C. Salles: Data curation, investigation. S. Murali: Data curation, investigation. J.L. Hicks: Methodology. M. Nguyen: Methodology. C.C. Pritchard: Conceptualization, data curation, investigation. A.M. De Marzo: Conceptualization, data curation, investigation. J.S. Lanchbury: Data curation, funding acquisition, investigation, methodology. B.J. Trock: Data curation, visualization, writing-review and editing. W.B. Isaacs: Conceptualization, data curation, investigation, writing-original draft, writing-review and editing. K.M. Timms: Conceptualization, data curation, formal analysis, investigation, writing-original draft, writing-review and editing. E.S. Antonarakis: Conceptualization, resources, data curation, formal analysis, funding acquisition, investigation, writing-original draft, project administration, writing-review and editing. T.L. Lotan: Conceptualization, data curation, formal analysis, supervision, validation, investigation, writing-original draft, project administration, writing-review and editing.

This work was supported by the Patrick Walsh Prostate Cancer Research Fund (to E.S. Antonarakis), the Prostate Cancer Foundation (to E.S. Antonarakis), NIH/NCI Prostate SPORE P50CA58236 (to A.M. De Marzo), and the NCI Cancer Center Support Grant 5P30CA006973-52 (to T.L. Lotan and E.S. Antonarakis) as well as the NIH/NCI U01 CA196390 (to A.M. De Marzo) and the U.S. Department of Defense Prostate Cancer Research Program Prostate Cancer Biospecimen Network Site (W81XWH-18-2-0015 to A.M. De Marzo).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Shiloh
Y
,
Ziv
Y
. 
The ATM protein kinase: regulating the cellular response to genotoxic stress, and more
.
Nat Rev Mol Cell Biol
2013
;
14
:
197
210
.
2.
Choi
M
,
Kipps
T
,
Kurzrock
R
. 
ATM mutations in cancer: therapeutic implications
.
Mol Cancer Ther
2016
;
15
:
1781
91
.
3.
Pritchard
CC
,
Mateo
J
,
Walsh
MF
,
De Sarkar
N
,
Abida
W
,
Beltran
H
, et al
Inherited DNA-repair gene mutations in men with metastatic prostate cancer
.
N Engl J Med
2016
;
375
:
443
53
.
4.
Robinson
D
,
Van Allen
EM
,
Wu
YM
,
Schultz
N
,
Lonigro
RJ
,
Mosquera
JM
, et al
Integrative clinical genomics of advanced prostate cancer
.
Cell
2015
;
161
:
1215
28
.
5.
Abida
W
,
Cyrta
J
,
Heller
G
,
Prandi
D
,
Armenia
J
,
Coleman
I
, et al
Genomic correlates of clinical outcome in advanced prostate cancer
.
Proc Natl Acad Sci U S A
2019
;
116
:
11428
36
.
6.
Na
R
,
Zheng
SL
,
Han
M
,
Yu
H
,
Jiang
D
,
Shah
S
, et al
Germline mutations in ATM and BRCA1/2 distinguish risk for lethal and indolent prostate cancer and are associated with early age at death
.
Eur Urol
2017
;
71
:
740
7
.
7.
Wu
Y
,
Yu
H
,
Li
S
,
Wiley
K
,
Zheng
SL
,
LaDuca
H
, et al
Rare germline pathogenic mutations of DNA repair genes are most strongly associated with grade group 5 prostate cancer
.
Eur Urol Oncol
2020
;
3
:
224
30
.
8.
Mateo
J
,
Boysen
G
,
Barbieri
CE
,
Bryant
HE
,
Castro
E
,
Nelson
PS
, et al
DNA repair in prostate cancer: biology and clinical implications
.
Eur Urol
2017
;
71
:
417
25
.
9.
Mateo
J
,
Porta
N
,
Bianchini
D
,
McGovern
U
,
Elliott
T
,
Jones
R
, et al
Olaparib in patients with metastatic castration-resistant prostate cancer with DNA repair gene aberrations (TOPARP-B): a multicentre, open-label, randomised, phase 2 trial
.
Lancet Oncol
2020
;
21
:
162
74
.
10.
Abida
W
,
Campbell
D
,
Patnaik
A
,
Sautois
B
,
Shapiro
J
,
Vogelzang
NJ
, et al
Preliminary results from the TRITON2 study of rucaparib in patients (pts) with DNA damage repair (DDR)-deficient metastatic castration-resistant prostate cancer (mCRPC): updated analyses
.
Ann Oncol
2019
;
30
:
327
.
11.
Smith
MR
,
Sandhu
SK
,
Kelly
WK
,
Scher
HI
,
Efstathiou
E
,
Lara
N
, et al
Pre-specified interim analysis of GALAHAD: a phase II study of niraparib in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC) and biallelic DNA-repair gene defects (DRD)
.
Ann Oncol
2019
;
30
:
884
.
12.
Marshall
CH
,
Sokolova
AO
,
McNatty
AL
,
Cheng
HH
,
Eisenberger
MA
,
Bryce
AH
, et al
Differential response to olaparib treatment among men with metastatic castration-resistant prostate cancer harboring BRCA1 or BRCA2 versus atm mutations
.
Eur Urol
2019
;
76
:
452
8
.
13.
Bono
JSD
,
Mehra
N
,
Higano
CS
,
Saad
F
,
Buttigliero
C
,
Mata
M
, et al
TALAPRO-1: A phase II study of talazoparib (TALA) in men with DNA damage repair mutations (DDRmut) and metastatic castration-resistant prostate cancer (mCRPC)—first interim analysis (IA)
.
J Clin Oncol
2020
;
38
:
119
.
14.
de Bono
J
,
Mateo
J
,
Fizazi
K
,
Saad
F
,
Shore
N
,
Sandhu
S
, et al
Olaparib for metastatic castration-resistant prostate cancer
.
N Engl J Med
2020
;
382
:
2091
102
.
15.
Luo
J
,
Antonarakis
ES
. 
PARP inhibition - not all gene mutations are created equal
.
Nat Rev Urol
2019
;
16
:
4
6
.
16.
Rafiei
S
,
Fitzpatrick
K
,
Liu
D
,
Cai
MY
,
Elmarakeby
HA
,
Park
J
, et al
ATM loss confers greater sensitivity to ATR inhibition than PARP inhibition in prostate cancer
.
Cancer Res
2020
;
80
:
2094
100
.
17.
Karnitz
LM
. 
Pathways ZLM: targeting ATR in cancer therapy
.
Clin Cancer Res
2015
;
21
:
4780
5
.
18.
Charrier
JD
,
Durrant
SJ
,
Golec
JM
,
Kay
DP
,
Knegtel
RM
,
MacCormick
S
, et al
Discovery of potent and selective inhibitors of ataxia telangiectasia mutated and Rad3 related (ATR) protein kinase as potential anticancer agents
.
J Med Chem
2011
;
54
:
2320
30
.
19.
Reaper
PM
,
Griffiths
MR
,
Long
JM
,
Charrier
JD
,
Maccormick
S
,
Charlton
PA
, et al
Selective killing of ATM- or p53-deficient cancer cells through inhibition of ATR
.
Nat Chem Biol
2011
;
7
:
428
30
.
20.
Bono
JSD
,
Tan
DSP
,
Caldwell
R
,
Terbuch
A
,
Goh
BC
,
Heong
V
, et al
First-in-human trial of the oral ataxia telangiectasia and Rad3-related (ATR) inhibitor BAY 1895344 in patients (pts) with advanced solid tumors
.
J Clin Oncol
2019
;
37
:
3007
.
21.
Antonarakis
ES
,
Piulats
JM
,
Gross-Goupil
M
,
Goh
J
,
Ojamaa
K
,
Hoimes
CJ
, et al
Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study
.
J Clin Oncol
2020
;
38
:
395
405
.
22.
Boudadi
K
,
Suzman
DL
,
Anagnostou
V
,
Fu
W
,
Luber
B
,
Wang
H
, et al
Ipilimumab plus nivolumab and DNA-repair defects in AR-V7-expressing metastatic prostate cancer
.
Oncotarget
2018
;
9
:
28561
71
.
23.
Isaacsson Velho
P
,
Qazi
F
,
Hassan
S
,
Carducci
MA
,
Denmeade
SR
,
Markowski
MC
, et al
Efficacy of radium-223 in bone-metastatic castration-resistant prostate cancer with and without homologous repair gene defects
.
Eur Urol
2019
;
76
:
170
6
.
24.
Teply
BA
,
Kachhap
S
,
Eisenberger
MA
,
Denmeade
SR
. 
Extreme response to high-dose testosterone in BRCA2- and ATM-mutated prostate cancer
.
Eur Urol
2017
;
71
:
499
.
25.
Chatterjee
P
,
Schweizer
MT
,
Lucas
JM
,
Coleman
I
,
Nyquist
MD
,
Frank
SB
, et al
Supraphysiological androgens suppress prostate cancer growth through androgen receptor-mediated DNA damage
.
J Clin Invest
2019
;
130
:
4245
60
.
26.
Mateo
J
,
Seed
G
,
Bertan
C
,
Rescigno
P
,
Dolling
D
,
Figueiredo
I
, et al
Genomics of lethal prostate cancer at diagnosis and castration resistance
.
J Clin Invest
2020
;
130
:
1743
51
.
27.
Velho
PI
,
Lim
D
,
Wang
H
,
Park
JC
,
Kaur
HB
,
Almutairi
F
, et al
Molecular characterization and clinical outcomes of primary Gleason pattern 5 prostate cancer after radical prostatectomy
.
JCO Precis Oncol
2019
;
3
:
1
13
.
28.
Lotan
TL
,
Torres
A
,
Zhang
M
,
Tosoian
JJ
,
Guedes
LB
,
Fedor
H
, et al
Somatic molecular subtyping of prostate tumors from HOXB13 G84E carriers
.
Oncotarget
2017
;
8
:
22772
82
.
29.
Chuang
AY
,
DeMarzo
AM
,
Veltri
RW
,
Sharma
RB
,
Bieberich
CJ
,
Epstein
JI
. 
Immunohistochemical differentiation of high-grade prostate carcinoma from urothelial carcinoma
.
Am J Surg Pathol
2007
;
31
:
1246
55
.
30.
Kaur
HB
,
Lu
J
,
Guedes
LB
,
Maldonado
L
,
Reitz
L
,
Barber
JR
, et al
TP53 missense mutation is associated with increased tumor-infiltrating T cells in primary prostate cancer
.
Hum Pathol
2019
;
87
:
95
102
.
31.
Tosoian
JJ
,
Almutairi
F
,
Morais
CL
,
Glavaris
S
,
Hicks
J
,
Sundi
D
, et al
Prevalence and prognostic significance of PTEN loss in African-American and European-American men undergoing radical prostatectomy
.
Eur Urol
2017
;
71
:
697
700
.
32.
Maughan
BL
,
Guedes
LB
,
Boucher
K
,
Rajoria
G
,
Liu
Z
,
Klimek
S
, et al
p53 status in the primary tumor predicts efficacy of subsequent abiraterone and enzalutamide in castration-resistant prostate cancer
.
Prostate Cancer Prostatic Dis
2018
;
21
:
260
8
.
33.
Ross
AE
,
Johnson
MH
,
Yousefi
K
,
Davicioni
E
,
Netto
GJ
,
Marchionni
L
, et al
Tissue-based genomics augments post-prostatectomy risk stratification in a natural history cohort of intermediate- and high-risk men
.
Eur Urol
2016
;
69
:
157
65
.
34.
Johnson
MH
,
Ross
AE
,
Alshalalfa
M
,
Erho
N
,
Yousefi
K
,
Glavaris
S
, et al
SPINK1 defines a molecular subtype of prostate cancer in men with more rapid progression in an at risk, natural history radical prostatectomy cohort
.
J Urol
2016
;
196
:
1436
44
.
35.
Miller
RM
,
Nworu
C
,
McKee
L
,
Balcerzak
D
,
Pham
L
,
Pugh
J
, et al
Development of an immunohistochemical assay to detect the ataxia-telangiectasia mutated (ATM) protein in gastric carcinoma
.
Appl Immunohistochem Mol Morphol
2020
;
28
:
303
10
.
36.
Bang
YJ
,
Xu
RH
,
Chin
K
,
Lee
KW
,
Park
SH
,
Rha
SY
, et al
Olaparib in combination with paclitaxel in patients with advanced gastric cancer who have progressed following first-line therapy (GOLD): a double-blind, randomised, placebo-controlled, phase 3 trial
.
Lancet Oncol
2017
;
18
:
1637
51
.
37.
Ahearn
TU
,
Pettersson
A
,
Ebot
EM
,
Gerke
T
,
Graff
RE
,
Morais
CL
, et al
A prospective investigation of PTEN loss and ERG expression in lethal prostate cancer
.
J Natl Cancer Inst
2016
;
108
:
djv346
.
38.
Chaux
A
,
Albadine
R
,
Toubaji
A
,
Hicks
J
,
Meeker
A
,
Platz
EA
, et al
Immunohistochemistry for ERG expression as a surrogate for TMPRSS2-ERG fusion detection in prostatic adenocarcinomas
.
Am J Surg Pathol
2011
;
35
:
1014
20
.
39.
Nguyen
M
,
Simmons
AD
,
Harding
TC
. 
Abstract 2476: Preclinical assessment of the PARP inhibitor rucaparib in homologous recombination deficient prostate cancer models
.
Cancer Res
2017
;
77
:
2476
.
40.
Lotan
TL
,
Gurel
B
,
Sutcliffe
S
,
Esopi
D
,
Liu
W
,
Xu
J
, et al
PTEN protein loss by immunostaining: analytic validation and prognostic indicator for a high risk surgical cohort of prostate cancer patients
.
Clin Cancer Res
2011
;
17
:
6563
73
.
41.
Patel
JN
,
Braicu
I
,
Timms
KM
,
Solimeno
C
,
Tshiaba
P
,
Reid
J
, et al
Characterisation of homologous recombination deficiency in paired primary and recurrent high-grade serous ovarian cancer
.
Br J Cancer
2018
;
119
:
1060
6
.
42.
Li
MM
,
Datto
M
,
Duncavage
EJ
,
Kulkarni
S
,
Lindeman
NI
,
Roy
S
, et al
Standards and guidelines for the interpretation and reporting of sequence variants in cancer: a joint consensus recommendation of the association for molecular pathology, American Society of Clinical Oncology, and College of American Pathologists
.
J Mol Diagn
2017
;
19
:
4
23
.
43.
Zhang
X
,
Loberiza
FR
,
Klein
JP
,
Zhang
MJ
. 
A SAS macro for estimation of direct adjusted survival curves based on a stratified cox regression model
.
Comput Methods Programs Biomed
2007
;
88
:
95
101
.
44.
Klein
JP
. 
Semiparametric estimation of random effects using the Cox model based on the EM algorithm
.
Biometrics
1992
;
48
:
795
806
.
45.
Sutton
LA
,
Ljungstrom
V
,
Mansouri
L
,
Young
E
,
Cortese
D
,
Navrkalova
V
, et al
Targeted next-generation sequencing in chronic lymphocytic leukemia: a high-throughput yet tailored approach will facilitate implementation in a clinical setting
.
Haematologica
2015
;
100
:
370
6
.
46.
Tomlins
SA
,
Rhodes
DR
,
Perner
S
,
Dhanasekaran
SM
,
Mehra
R
,
Sun
XW
, et al
Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer
.
Science
2005
;
310
:
644
8
.
47.
Cerveira
N
,
Ribeiro
FR
,
Peixoto
A
,
Costa
V
,
Henrique
R
,
Jeronimo
C
, et al
TMPRSS2-ERG gene fusion causing ERG overexpression precedes chromosome copy number changes in prostate carcinomas and paired HGPIN lesions
.
Neoplasia
2006
;
8
:
826
32
.
48.
Perner
S
,
Mosquera
JM
,
Demichelis
F
,
Hofer
MD
,
Paris
PL
,
Simko
J
, et al
TMPRSS2-ERG fusion prostate cancer: an early molecular event associated with invasion
.
Am J Surg Pathol
2007
;
31
:
882
8
.
49.
Morais
CL
,
Guedes
LB
,
Hicks
J
,
Baras
AS
,
De Marzo
AM
,
Lotan
TL
. 
ERG and PTEN status of isolated high-grade PIN occurring in cystoprostatectomy specimens without invasive prostatic adenocarcinoma
.
Hum Pathol
2016
;
55
:
117
25
.
50.
Bismar
TA
,
Yoshimoto
M
,
Vollmer
RT
,
Duan
Q
,
Firszt
M
,
Corcos
J
, et al
PTEN genomic deletion is an early event associated with ERG gene rearrangements in prostate cancer
.
BJU Int
2011
;
107
:
477
85
.
51.
Gumuskaya
B
,
Gurel
B
,
Fedor
H
,
Tan
HL
,
Weier
CA
,
Hicks
JL
, et al
Assessing the order of critical alterations in prostate cancer development and progression by IHC: further evidence that PTEN loss occurs subsequent to ERG gene fusion
.
Prostate Cancer Prostatic Dis
2013
;
16
:
209
15
.
52.
De Marzo
AM
,
Haffner
MC
,
Lotan
TL
,
Yegnasubramanian
S
,
Nelson
WG
. 
Premalignancy in prostate cancer: rethinking what we know
.
Cancer Prev Res
2016
;
9
:
648
56
.
53.
Cancer Genome Atlas Research Network
. 
The molecular taxonomy of primary prostate cancer
.
Cell
2015
;
163
:
1011
25
.
54.
Castro
E
,
Romero-Laorden
N
,
Del Pozo
A
,
Lozano
R
,
Medina
A
,
Puente
J
, et al
PROREPAIR-B: a prospective cohort study of the impact of germline DNA repair mutations on the outcomes of patients with metastatic castration-resistant prostate cancer
.
J Clin Oncol
2019
;
37
:
490
503
.
55.
Antonarakis
ES
,
Lu
C
,
Luber
B
,
Liang
C
,
Wang
H
,
Chen
Y
, et al
Germline DNA-repair gene mutations and outcomes in men with metastatic castration-resistant prostate cancer receiving first-line abiraterone and enzalutamide
.
Eur Urol
2018
;
74
:
218
25
.
56.
Annala
M
,
Struss
WJ
,
Warner
EW
,
Beja
K
,
Vandekerkhove
G
,
Wong
A
, et al
Treatment outcomes and tumor loss of heterozygosity in germline DNA repair-deficient prostate cancer
.
Eur Urol
2017
;
72
:
34
42
.
57.
Castro
E
,
Goh
C
,
Olmos
D
,
Saunders
E
,
Leongamornlert
D
,
Tymrakiewicz
M
, et al
Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer
.
J Clin Oncol
2013
;
31
:
1748
57
.
58.
Carter
HB
,
Helfand
B
,
Mamawala
M
,
Wu
Y
,
Landis
P
,
Yu
H
, et al
Germline mutations in ATM and BRCA1/2 are associated with grade reclassification in men on active surveillance for prostate cancer
.
Eur Urol
2019
;
75
:
743
9
.
59.
Schweizer
MT
,
Antonarakis
ES
,
Bismar
TA
,
Guedes
LB
,
Cheng
HH
,
Tretiakova
MS
, et al
Genomic characterization of prostatic ductal adenocarcinoma identifies a high prevalence of DNA repair gene mutations
.
JCO Precis Oncol
2019
;
3
:
1
9
.
60.
Marshall
CH
,
Fu
W
,
Wang
H
,
Baras
AS
,
Lotan
TL
,
Antonarakis
ES
. 
Prevalence of DNA repair gene mutations in localized prostate cancer according to clinical and pathologic features: association of Gleason score and tumor stage
.
Prostate Cancer Prostatic Dis
2018
;
22
:
59
65
.
61.
Abida
W
,
Campbell
D
,
Patnaik
A
,
Shapiro
JD
,
Sautois
B
,
Vogelzang
NJ
, et al
Non-BRCA DNA damage repair gene alterations and response to the PARP inhibitor rucaparib in metastatic castration-resistant prostate cancer: analysis from the phase II TRITON2 study
.
Clin Cancer Res
2020
;
26
:
2487
96
.
62.
Trotman
LC
,
Niki
M
,
Dotan
ZA
,
Koutcher
JA
,
Di Cristofano
A
,
Xiao
A
, et al
Pten dose dictates cancer progression in the prostate
.
PLoS Biol
2003
;
1
:
E59
.
63.
Lu
S
,
Shen
K
,
Wang
Y
,
Santner
SJ
,
Chen
J
,
Brooks
SC
, et al
Atm-haploinsufficiency enhances susceptibility to carcinogen-induced mammary tumors
.
Carcinogenesis
2006
;
27
:
848
55
.
64.
Umesako
S
,
Fujisawa
K
,
Iiga
S
,
Mori
N
,
Takahashi
M
,
Hong
DP
, et al
Atm heterozygous deficiency enhances development of mammary carcinomas in p53 heterozygous knockout mice
.
Breast Cancer Res
2005
;
7
:
R164
70
.
65.
Ahuja
AK
,
Barber
RC
,
Hardwick
RJ
,
Weil
MM
,
Genik
PC
,
Brenner
DJ
, et al
The effects of Atm haploinsufficiency on mutation rate in the mouse germ line and somatic tissue
.
Mutagenesis
2008
;
23
:
367
70
.
66.
Connolly
L
,
Lasarev
M
,
Jordan
R
,
Schwartz
JL
,
Turker
MS
. 
Atm haploinsufficiency does not affect ionizing radiation mutagenesis in solid mouse tissues
.
Radiat Res
2006
;
166
:
39
46
.
67.
Abida
W
,
Armenia
J
,
Gopalan
A
,
Brennan
R
,
Walsh
M
,
Barron
D
, et al
Prospective genomic profiling of prostate cancer across disease states reveals germline and somatic alterations that may affect clinical decision making
.
JCO Precis Oncol
2017
;
1
:
1
16
.
68.
Pritchard
CC
,
Morrissey
C
,
Kumar
A
,
Zhang
X
,
Smith
C
,
Coleman
I
, et al
Complex MSH2 and MSH6 mutations in hypermutated microsatellite unstable advanced prostate cancer
.
Nat Commun
2014
;
5
:
4988
.