Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 May 3.
Published in final edited form as: Clin Cancer Res. 2020 Oct 7;26(24):6505–6512. doi: 10.1158/1078-0432.CCR-20-1788

Effect of germline mutations in homologous recombination repair genes on overall survival of patients with pancreatic adenocarcinoma

Siddhartha Yadav 1,+, Pashtoon M Kasi 2,+, William R Bamlet 4, Thanh P Ho 1, Eric C Polley 3,4, Chunling Hu 3, Steven N Hart 4, Kari G Rabe 4, Nicholas J Boddicker 4, Rohan D Gnanaolivu 4, Kun Y Lee 3, Tricia H Lindstrom 4, Gloria M Petersen 4, Fergus J Couch 3, Robert R McWilliams 1
PMCID: PMC9063708  NIHMSID: NIHMS1635289  PMID: 33028596

Abstract

Purpose:

To compare the clinical characteristics and overall survival (OS) of germline mutation carriers in homologous recombination repair (HRR) genes and non-carriers with pancreatic ductal adenocarcinoma (PDAC).

Methods:

Germline DNA from 3,078 patients with PDAC enrolled in a prospective registry at Mayo Clinic between 2000 and 2017 was analyzed for mutations in 37 cancer predisposition genes. Characteristics and OS of patients with mutations in 8 genes (ATM, BARD1, BRCA1, BRCA2, BRIP1, PALB2, RAD51C, and RAD51D) involved in HRR were compared to patients testing negative for mutations in all 37 genes.

Results

The 175 HRR mutation carriers and 2,730 non-carriers in the study had a median duration of follow-up of 9.9 years. HRR mutation carriers were younger (Median age at diagnosis: 63 vs. 66 years, p<0.001) and more likely to have metastatic disease at diagnosis (46% vs. 36%, p=0.004). In a multivariable model adjusting for sex, age at diagnosis, and tumor staging, patients with germline HRR mutations had a significantly longer OS compared to non-carriers (HR: 0.83, 95% CI: 0.70 to 0.97, p= 0.02). Further gene-level analysis demonstrated that germline ATM mutation carriers had longer OS compared to patients without germline mutations in any of the 37 HRR genes (HR: 0.72, 95% CI: 0.55 – 0.94, p=0.01).

Conclusions

This study demonstrates that germline mutation carrier status in PDAC is associated with longer OS compared to non-carriers. Further research into tumor biology and response to platinum-based chemotherapy in germline mutation carriers with PDAC are needed to better understand the association with longer OS.

Keywords: Overall survival, pancreatic cancer, BRCA, Homologous recombination repair, Germline, Mutation

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a five-year survival of less than 10% (1, 2). There is an ongoing effort to improve the overall survival of PDAC by identifying potentially targetable somatic or germline mutations. In recent years, it has been demonstrated that germline mutations in several cancer predisposition genes may confer an increased risk of PDAC, and are detected in approximately 5–15% of patients with PDAC (36). Some studies have also demonstrated that the biological characteristics of PDAC tumors in germline mutation carriers of homologous recombination repair (HRR) are different from sporadic PDACs (7, 8). In addition, germline mutations in HRR genes are known to confer higher sensitivity to DNA-damaging agents and/or ionizing radiation (4, 911). Furthermore, a new oral class of therapeutic agents called poly-adenosine diphosphate-ribose polymerase (PARP) inhibitors has been developed to take advantage of impaired HRR in tumor cells to induce synthetic lethality. Recently, maintenance therapy with a PARP inhibitor was shown to improve progression free survival in patients with germline BRCA1 or BRCA2 (BRCA1/2 hereafter) mutations and metastatic PDAC compared to placebo (12).

Despite the improved understanding of differences in tumor biology and response to therapy in mutation carriers of HRR genes, the impact of mutation status on overall survival (OS) is still unclear. Prior studies on the impact of germline mutations on OS in PDAC had demonstrated mixed results, but these were mostly restricted to BRCA1/2 mutations and were limited by small sample sizes (1316). Since several genes involved in HRR have similar biological functions, the differences in tumor biology and response to therapy may not be limited to BRCA1/2 mutation carriers. In this context, we evaluated the clinical characteristics of PDAC in germline mutation carriers of HRR genes and implications of these mutations on OS in large prospective hospital-based cohort of patients with PDAC.

Materials and Methods

Patient selection and Follow-up:

The study sample was derived from the Mayo Clinic Biospecimen Resource for Pancreas Research, a prospective registry offering participation to patients evaluated at Mayo Clinic Rochester for initial diagnosis of pancreatic cancer. Patients enrolled in this registry between October 12, 2000 and October 6, 2017 with PDAC were included. Patients who were enrolled in the registry more than 3 months after their initial diagnosis and those who had no further follow-up after their diagnosis were excluded. While the results of germline sequencing have previously been reported (3), the present analysis focused on OS and included 454 additional cases enrolled after the prior publication. This study was approved by the Institutional Review Board (IRB) at Mayo Clinic, and all aspects of the study were in accordance with the Declaration of Helsinki. . All patients signed written, informed consent.

Patients consenting to the study completed a baseline questionnaire on personal and family history and provided a blood sample. In addition, patient demographics, tumor characteristics, family history, and clinical outcomes were abstracted from electronic medical records to verify existing information reported by patients. For patients who did not follow up at Mayo Clinic, medical records from local institutions were requested at 1 year post diagnosis, 3 years post diagnosis and at death. A medical oncologist or trained nurse abstractor reviewed each record for abstraction of information pertaining to any treatment the patient received. Furthermore, additional follow up questionnaires were sent to patients to obtain information on treatment and outcomes while participating in the study. The abstraction of information on treatment was complete on approximately two-thirds (64.4%) of the study participants at the time of last follow up or death. For the purpose of this study, patients treated with cisplatin, oxaliplatin or carboplatin at any point during the course of treatment were considered to have received platinum-based chemotherapy.

Germline sequencing and Bioinformatics Analysis:

Germline DNA extracted from peripheral blood mononuclear cells was analyzed for germline pathogenic variants in the coding regions and consensus splice sites of 37 genes (Supplemental information and Supplemental Table 1) using a custom amplicon-based QIAseq panel (QIAGEN, Hilden, Germany). Pooled sample libraries from 768 samples were subjected to paired-end 150-base pair sequencing in each lane of a HiSeq4000 (Illumina, San Diego, CA) with a median coverage of 200X. Reads were trimmed with Cutadapt version 1.10(17) and aligned with BWA-MEM (18). Sequence realignment, recalibration, haplotype calling, and depth of coverage were conducted using Genome Analysis Toolkit (GATK) version 3.4–46 (19). Large genomic rearrangements were detected with Pattern CNV v1.1.3 (20). Annotation of mutations was provided through the BioR toolkit(21) leveraging dbNSFP v3.0(22), ClinVar (23), Clinical Annotation of Variants (24), and population frequencies from Genome Aggregation Database (gnomAD) (11) and Exome Aggregation Consortium (ExAC) (25). Pathogenic mutations were viewed with VCF-Miner (26). A 5-tier system was used to classify mutations based on the American College of Medical Genetics and the Association for Molecular Pathology guidelines (27). Pathogenic and likely pathogenic variants were analyzed together as pathogenic variants.

Statistical Analysis:

Patients with a germline mutation in one of the eight genes (ATM, BARD1, BRCA1, BRCA2, BRIP1, PALB2, RAD51C, and RAD51D) involved in HRR were considered as carriers whereas those who tested negative for germline mutation in all 37 genes were considered non-carriers. These 8 genes were selected based on prior studies demonstrating association with homologous recombination deficiency (28). Since other genes in the QIAseq panel may be associated with HRR, mutation carriers in genes other than the 8 HRR genes were excluded from primary analysis to mitigate their effect. Baseline characteristics between carriers and non-carriers were compared using Fisher’s exact test for categorical variables and ANOVA for continuous variables. OS between carriers and non-carriers was compared in a multivariable cox-proportional hazard regression model including age, sex, and stage at diagnosis as covariates along with subset analysis by specific chemotherapy regimens, year of diagnosis, and specific gene mutations. Separate sensitivity analyses for differences in baseline characteristics and OS were performed in patients with germline mutations in the 8 HRR genes (carriers) and in those who tested negative for mutations in the 8 HRR genes (non-carriers). All statistical tests were performed using SAS 9.4 statistical software (SAS Institute, Cary, NC).

Results

Out of the 3,078 patients included in the final analysis, 175 (5.7%) were found to carry a mutation in one of the 8 HRR genes: 67 (2.2%) in BRCA2, 65 (2.1%) in ATM, 20 (0.6%) in BRCA1, 12 (0.4%) in PALB2, 4 (0.1%) in BRIP1, 4 (0.1%) in RAD51C, 2 (0.1%) in BARD1, and 1 (0.03%) in RAD51D (Table 1). The non-carriers included 2730 (88.7%) patients without mutations in any of the 37 genes evaluated. The median age at diagnosis of the cohort was 65.8 years with a median duration of follow-up of 9.9 years.

Table 1:

Frequencies of germline mutations in the cohort

Gene Number of mutation carriers (N=3078) Percent
APC 2 0.1%
ATM 65 2.1%
BARD1 2 0.1%
BLM 9 0.3%
BRCA1 20 0.6%
BRCA2 67 2.2%
BRIP1 4 0.1%
CDKN2A 10 0.3%
CHEK2 22 0.7%
EPCAM 1 0.0%
ERCC2 13 0.4%
ERCC3 7 0.2%
FANCC 8 0.3%
FANCM 8 0.3%
KRAS 1 0.0%
MLH1 6 0.2%
MRE11A 2 0.1%
MSH2 1 0.0%
MSH6 9 0.3%
MUTYH 6 0.2%
NBN 4 0.1%
NF1 2 0.1%
PALB2 12 0.4%
PMS2 10 0.3%
PPM1D 23 0.7%
PRSS1 5 0.2%
RAD50 6 0.2%
RAD51C 4 0.1%
RAD51D 1 0.0%
RECQL 8 0.3%
RINT1 5 0.2%
SLX4 5 0.2%
TP53 6 0.2%
XRCC2 3 0.1%

Baseline Characteristics of Mutation Carriers and Non-Carriers:

Baseline characteristics of 175 mutation carriers in 8 HRR genes and 2,730 non-carriers are presented in Table 2. Mutation carriers were diagnosed with PDAC at a younger age (62.8 vs. 65.8 years, p<0.001) and were more likely to present with metastatic disease at diagnosis (46.2% vs. 35.6%, p<0.001) compared to non-carriers (Table 2). In addition, a higher proportion of mutation carriers had PDAC in the body or tail of the pancreas compared to non-carriers. Among patients with resectable PDAC, the rate of surgery was similar between the two groups. Furthermore, there were no statistically significant differences in the rates of chemotherapy between mutation carriers and non-carriers. The demographic and tumor characteristics of ATM carriers, which is one of the largest group of mutation carriers in this cohort is presented in supplemental table 2.

Table 2:

Baseline and clinical characteristics of mutation carriers of 8 HRR genes and non-carriers

Carrier (N=175) Non-Carrier (N=2730) P-value
Mean age at Diagnosis (SD) 62.78 (10.69) 65.82 (10.70) <0.001
Gender: 0.34
Male 106 (60.6%) 1553 (56.9%)
Female 69 (39.4%) 1177 (43.1%)
Ethnicity: 0.87
Non-Hispanic White 165 (95.9%) 2616 (96.2%)
Other 7 (4.1%) 104 (3.8%)
Missing 3 10
Mean BMI (SD) 28.74 (5.58) 28.59 (5.69) 0.51
Patient reported diabetes: 0.63
Yes 42 (24.0%) 700 (25.6%)
No 133 (76.0%) 2030 (74.4%)
Ever Smoker: 0.66
Yes 91 (55.2%) 1493 (56.9%)
No 74 (44.8%) 1132 (43.1%)
Missing 10 105
Mean pack years in ever smokers (SD) 26.00 (20.64) 28.18 (25.33) 0.82
Site of Pancreas Mass: 0.002
 Head and adjacent parts 94 (55.6%) 1789 (67.2%)
 Body/Tail 75 (44.4%) 872 (32.8%)
 Missing 6 69
Pancreas cancer stage grouping: 0.004
 Resectable 46 (26.6%) 692 (25.4%)
 Locally Advanced 47 (27.2%) 1059 (38.9%)
 Metastatic 80 (46.2%) 969 (35.6%)
 Missing 2 10
Pancreas cancer surgery: 0.62
 Pancreatico-duodenectomy 41 (71.9%) 611 (73.5%)
 Distal pancreatectomy 15 (26.3%) 188 (22.6%)
 Total pancreatectomy 1 (1.8%) 32 (3.9%)
 No-surgery or missing 118 1899
Chemotherapy: 0.56
 Yes 119 (83.8%) 1590 (81.9%)
 No Chemotherapy 23 (16.2%) 352 (18.1%)
 Missing 33 788
Radiation: 0.03
 Yes 64 (50.0%) 769 (40.0%)
 No radiation 64 (50.0%) 1152 (60.0%)
 Missing 47 809

BMI: Body Mass Index; SD: Standard Deviation

Overall Survival:

The median OS for mutation carriers was 14.6 months (95% CI: 13.2 – 16.1 months) compared to 11.7 months (95% CI: 11.2 – 12.4 months) for non-carriers (Figure 1). In univariate analysis, the difference in the OS between mutation carriers and non-carriers did not reach statistical significance (HR: 0.86, 95% CI: 0.73 – 1.01, p= 0.07). However, in multivariable analysis adjusting for age, sex, and stage at diagnosis, mutation carrier status was associated with a significantly longer OS compared to non-carriers (HR: 0.83, 95% CI: 0.70 – 0.97, p= 0.02) (Table 3). Among surgically resectable patients, the median OS was 23.7 months in mutation carriers and 23.5 months in non-carriers (Supplemental Figure 1) whereas among non-resectable/metastatic patients, the median OS was 11.5 months in mutation carriers and 9.6 months in non-carriers (Supplemental Figure 2).

Figure 1:

Figure 1:

Kaplan-Meier curves for overall survival in homologous recombination repair gene mutation carriers and non-carriers

Table 3:

Multivariable analysis for overall survival comparing the 8 HRR gene mutation carriers to non-carriers#.

Number of Events/Total Median Survival in months (95% CI) Multivariable Analysis
Hazard Ratio (95% CI) P-value
Overall: 0.02
 Non-Carrier 2441/2730 11.7 (11.2–12.4) Reference
 Carrier 154/175 14.6 (13.2–16.1) 0.83 (0.70–0.97)
Subset Analysis:
Non-resectable or metastatic 0.02
 Non-Carrier 1839/2028 9.6 (9.1 – 10.2) Reference
 Carrier 118/127 8.0 (5.8–12.2) 0.81 (0.67 – 0.97)
Surgically resected 0.49
 Non-Carrier 565/692 23.5 (21.7 – 25.2) Reference
 Carrier 34/46 23.7 (18.4 – 45.8) 0.89 (0.63 – 1.25)
Diagnosed prior to May 2011 0.22
 Non-Carrier 1650/1741 10.4 (9.7–11.0) Reference
 Carrier 105/114 13.8 (10.6–16.1) 0.89 (0.73–1.08)
Diagnosed May 2011 or later 0.04
 Non-Carrier 791/989 13.7 (13.1–14.3) Reference
 Carrier 49/61 15.7 (12.4–20.5) 0.75 (0.56–1.00)
Received platinum-based chemotherapy 0.18
 Non-Carrier 597/700 16.3 (15.6–17.2) Reference
 Carrier 54/61 17.8 (16.0–22.1) 0.83 (0.63–1.10)
Did not receive platinum-based chemotherapy 0.20
 Non-Carrier 1838/2022 9.7 (9.1 – 10.2) Reference
 Carrier 98/112 11.5 (8.3 – 14.0) 0.88 (0.72 – 1.08)
Diagnosed prior to May 2011 and did not receive platinum-based chemotherapy 0.34
 Non-Carrier 1402/1485 9.3 (8.6 – 9.9) Reference
 Carrier 81/89 10.1 (8.0 – 14.0) 0.90 (0.72 – 1.13)
Diagnosed prior to May 2011 and received platinum-based chemotherapy 0.43
 Non-Carrier 243/249 15.6 (14.5–17.4) Reference
 Carrier 22/23 17.8 (15.0–45.8) 0.84 (0.54–1.31)
Diagnosed May 2011 or later & received platinum-based chemotherapy 0.24
 Non-Carrier 354/451 16.7 (15.9–17.5) Reference
 Carrier 32/38 19.7 (15.4–22.3) 0.81 (0.56–1.17)
Diagnosed May 2011 or later and received FOLFIRINOX 0.43
 Non-Carrier 246/322 17.4 (16.6–20.0) Reference
 Carrier 28/32 17.7 (12.4–28.5) 0.85 (0.57–1.27)
Diagnosed May 2011 or later and received irinotecan 0.41
 Non-Carrier 291/382 17.3 (16.6–19.3) Reference
 Carrier 30/35 17.7 (12.4–25.0) 0.85 (0.58–1.25)
Non-resectable/metastatic and received platinum-based chemotherapy 0.12
 Non-Carrier 490/591 15.4 (14.1 – 16.6) Reference
 Carrier 42/47 17.5 (14.6 – 21.8) 0.78 (0.57 – 1.07)
Surgically resected and received platinum-based chemotherapy 0.86
 Non-Carrier 106/107 22.9 (20.1 – 27.1) Reference
 Carrier 12/14 23.7 (17.7 – NE) 1.06 (0.57 – 1.95)
Subset analysis by mutation carrier status in each gene
 Non-carriers 2441/2730 11.7 (11.2–12.4) Reference
ATM carriers 57/65 15.7 (14.4–21.1) 0.72 (0.55–0.94) 0.01
BRCA1 carriers 19/20 9.2 (5.8–16.1) 1.26 (0.80–1.98) 0.34
BRCA2 carriers 58/67 15.0 (11.5–20.0) 0.81 (0.62–1.05) 0.10
PALB2 carriers 10/12 14.2 (12.4–66.9) 0.74 (0.40–1.38) 0.33
Subset analysis by receipt of platinum-based chemotherapy among mutation carriers
 Did not receive platinum-based chemotherapy 76/80 7.7 (5.8 – 10.1) Reference
 Received platinum-based chemotherapy 42/47 17.5 (14.6 – 21.8) 0.46 (0.30 – 0.68) <0.001

CI: Confidence Interval; FOLFIRINOX: Regimen consisting of fluorouracil, leucovorin, irinotecan and oxaliplatin;

#

: Analyses were adjusted for age at diagnosis, sex, and stage at diagnosis (where appropriate);

: Survival in carriers of germline mutations in each of the 8 homologous recombination repair genes compared to non-carriers; Hazard ratios for genes with less than 5 mutation carriers not estimated.

NE not estimable

Subgroup Analyses:

Further exploratory analyses were performed by stage of diagnosis, year of diagnosis, and type of chemotherapy (Table 3). Among patients with metastatic or non-resectable PDAC, mutation carriers were observed to have a significantly longer OS compared to non-carriers (HR: 0.81, 95%CI: 0.67 – 0.97, p=0.02). In addition, mutation carrier status was also significantly associated with longer OS among patients diagnosed after May 2011 (when FOLFIRINOX use started) compared to non-carriers diagnosed during the same time period (HR: 0.75, 95% CI: 0.56 – 1.00, p=0.04). However, specific chemotherapy regimens including platinum agents and FOLFIRINOX were not associated with OS. Similarly, on separate analysis of metastatic/non-resectable and surgically-resectable PDAC cases receiving platinum-based chemotherapy, mutation carriers were not noted have a statistically significant difference in OS compared to non-carriers (Table 3). Furthermore, no difference in OS by mutation carrier status was noted among patients not receiving platinum-based chemotherapy. However, restricting the analysis to mutation carriers with metastatic or non-resectable disease, patients who received platinum-based chemotherapy had better OS compared to mutation carriers who did not receive platinum based chemotherapy (HR:0.46, 95%CI:0.30 – 0.68, p<0.001) (Supplemental Figure 3).

Further gene-level analysis demonstrated that germline ATM mutation carriers had longer OS compared to patients without germline mutations in any of the 37 HRR genes (HR: 0.72, 95% CI: 0.55 – 0.94, p=0.01). Significant differences in OS were not noted for BRCA1, BRCA2, or PALB2 carriers compared to non-carriers (Table 3 and Supplemental Figures 45).

Sensitivity Analysis:

In sensitivity analysis including 175 mutation carriers in 8 HRR genes compared to the remaining 2,903 patients negative only for a mutation in these 8 genes, the overall differences in baseline characteristics and OS were similar to the primary analysis (Supplemental Tables 3 and 4).

Discussion

In a large prospective registry of patients with PDAC, we observed an association between germline HRR mutations and improved OS. Prior studies primarily focused on OS in PDAC with BRCA1/2 mutations and exposure to platinum based chemotherapy (13, 14, 2933). These studies have demonstrated mixed results, with some studies demonstrating longer OS (29, 33) while others demonstrated shorter or similar OS (13, 14). Fewer studies have evaluated the role of other DNA-damage repair or HRR genes (15, 3336). However, the majority of the prior studies were limited by their retrospective nature and small sample. In contrast, the present study is the largest to date, to the best of our knowledge, for carriers of HRR genes in patients with PDAC. In addition, the present study included a large number of non-carriers. Furthermore, the prospective collection of clinical outcomes is a significant strength of this study. The findings of our study have significant implications for future research, primarily in understanding the differences in tumor biology and response to standard treatments in germline mutation carriers with PDAC. In addition, these findings will aid in appropriate counseling of prognosis in mutation carriers with PDAC.

The difference in OS observed in this study could be due to differences in tumor biology and/or responses to standard treatments in germline mutation carriers with PDAC. Differences in OS due to unique tumor biology would be suggestive of prognostic nature of HRR mutations while OS difference due to response to therapy, e.g. platinum-based chemotherapy, would point towards predictive nature of HRR alterations. The prognostic versus predictive nature of HRR defect deserves special attention. Molecular studies involving sporadic PDAC have identified a complex mutational landscape with a high frequency of somatic mutations in KRAS, TP53, SMAD4, CDKN2A, and in genes involved in cell cycle regulation, TGF-β signaling, HRR, chromatin regulation, and axonal guidance pathways (7, 8, 3739). It is not entirely clear if germline mutation associated PDACs have similar landscapes of somatic mutations and mutational signatures as sporadic PDAC, but differences in tumor biology have been reported from smaller series. For example, significant enrichment for germline mutations in HRR genes in KRAS wild-type PDAC (8) and among patients with the mutational signature associated with high genomic instability (7) have been observed. While this study did not evaluate somatic mutations in PDAC, it does establish a foundation for future studies to evaluate if differences in tumor biology may explain the association with improved survival observed in this study.

Prior studies have demonstrated increased responsiveness to radiation and chemotherapy, particularly platinum-based chemotherapies, in PDAC associated with germline mutations (9, 14, 36, 40, 41). In addition, among patients with advanced PDAC treated with platinum based chemotherapy, OS has been shown to be longer for patients with HRR defect (33, 42, 43), which is suggestive of a predictive effect of HRR mutation on OS with platinum-based chemotherapy. While we observed that mutation carriers with advanced (non-resectable or metastatic) PDAC who received platinum-based chemotherapy had better OS compared to mutation carriers who did not (Supplemental Figure 1), we did not observe an association between OS and mutation carrier status among patients receiving platinum-based chemotherapy (Table 3). These findings highlight the importance of platinum-based chemotherapies in mutation carriers but do not confirm the predictive effect of HRR mutations on OS with platinum-based chemotherapy. Similarly, the association with improved OS in mutation carriers after platinum based therapy became standard of care in May 2011and in non-resectable/metastatic cases (in whom platinum-based chemotherapy is more likely to be administered) suggest, but do not confirm, that HRR defect may be predictive of differential response to platinum-based therapies. In contrast, the observation of no significant difference in OS between mutation carriers and non-carriers with specific chemotherapy regimens such as FOLFIRINOX argues against predictive effect of platinum-based chemotherapy. However, the lack of predictive effect of HRR mutation on OS with platinum-based chemotherapy in this study may be due to smaller numbers of patients in the analyzed subsets, and does not rule out this possibility.

Ultimately, this study is not adequately powered to delineate whether the improvement in OS in germline mutation carriers is due to higher response to chemotherapy or inherent differences in tumor biology or a combination of both. However, similar to other studies (4143), this study suggests that platinum-based therapies are important in treatment of mutation carriers with advanced PDAC. The current National Comprehensive Cancer Network guidelines on PDAC supports the use of cisplatin in BRCA1/2 mutation carriers, but the use of these agents in PDAC associated with other HRR genes has not been well-studied. Our study suggests that the sensitizing effects of platinum agents may extend beyond BRCA1/2 to other HRR genes. Larger randomized studies are needed to identify if mutation carriers in HRR genes other than BRCA1/2demonstrate significant responseto particular chemotherapy regimens. Biomarkers predictive of differential therapeutic response are also urgently needed. In addition, with the availability of PARP inhibitors, future clinical trials could include rational combinations of PARP-inhibitors with chemotherapeutic agents, immunotherapy and/or radiation therapy to leverage the germline mutation status to further improve OS (10).

The present study also demonstrated significant difference in OS between ATM mutation carriers and non-carriers. This is in contrast to a prior study that demonstrated poor survival with tumoral loss of ATM by immunohistochemistry in PDAC (44). The biological characteristics of PDAC tumors associated with germline ATM carriers are understudied. In a small study of 24 breast cancer tumors from germline ATM carriers, none of the tumors displayed high activity of mutational signature 3 associated with HRR defect (45). In addition, it has been noted that germline ATM mutations and TP53 somatic mutations may be epistatic (41, 45). Somatic mutation in TP53 is known to be associated with poor prognosis in multiple malignancies including PDAC (4648). These findings raise the possibility that germline ATM associated PDAC might be a distinct entity compared to BRCA1/2 associated PDAC or sporadic PDAC. Further evaluation of tumor biology of ATM associated PDAC tumors through DNA sequencing and RNA expression studies are needed to fully understand the unique nature of these tumors and implications on OS. In contrast, this study did note a difference in OS for BRCA1 or BRCA2. These analyses at gene-level may be underpowered to detect a difference in OS. However, further evaluation of differences in tumor biology and response to therapy between ATM, BRCA1/2 and PALB2 carriers should also be evaluated in larger studies in the future.

In addition to the survival differences, this study also showed that germline mutation carriers with PDAC are more likely to be younger and present with metastatic disease at diagnosis. Germline mutation carriers of HRR genes are diagnosed with breast and ovarian cancers at an earlier age (49) and are also more likely to have metastatic rather than localized prostate cancer compared to the general population (50). Similar associations in PDAC, although suspected, had not been observed previously and are novel findings of this study.

Limitations

Because this study was performed using a prospective registry from a single institution, it has several strengths compared with similar prior studies. However, a number of limitations exist. Since we only analyzed patients who consented to participate in the registries, a selection bias cannot be ruled out. We evaluated combined outcomes for 8 clinically relevant HRR genes, but it is possible that not all of the HRR genes may have similar biological properties. In addition, we did not take into account somatic mutations in tumors, which can independently affect outcomes as patients with somatic HRR defect are known to respond to platinum based chemotherapy and PARP inhibitors (41, 51, 52). Furthermore, the lack of ethnic/racial diversity and incomplete information on treatment is an important limitation of this study. Finally, even though we evaluated a large number of patients, the total number of germline mutation carriers was relatively small, and we may not have had power to detect differences in sub-group analyses.

Conclusions

This study demonstrates that germline mutation carriers with PDAC have a longer OS compared to non-carriers after adjusting for age, sex, and stage at diagnosis. Further research into tumor biology and therapeutic response to chemotherapy in PDAC associated with germline mutations in HRR genes are needed to identify not only prognostic but also predictive biomarkers in order to develop personalized treatment options for this unique patient population.

Supplementary Material

1

Translational Relevance of the study:

In this prospective study of patients with pancreatic cancer, an association between germline mutations in homologous recombination repair genes and improved overall survival was noted , possibly related to distinct tumor biology or increased response to therapy in mutation carriers.. Molecular studies involving sporadic pancreatic cancer have identified a complex mutational landscape with a high frequency of somatic mutations in KRAS, TP53, SMAD4, CDKN2A. However, it is not entirely clear if pancreatic tumors associated with germline mutations have similar landscapes of somatic mutations and mutational signatures as sporadic pancreatic cancer. In smaller studies, differences in tumor biology have been reported between sporadic pancreatic cancer and germline mutation associated pancreatic cancers. This study establishes a foundation for future studies to further investigate whether differences in tumor biology between sporadic and germline mutation associated pancreatic cancer may explain the association with improved survival observed in this study.

Acknowledgments:

This study was supported in part by NIH Specialized Program of Research Excellence in Pancreatic Cancer [CA102701], NIH Specialized Program of Research Excellence in Breast Cancer [CA116201], NIH grants CA116167, CA176785, CA192393, CA225662, the Breast Cancer Research Foundation and the Conquer Cancer Foundation Young Investigator Award .

RRM has served in consulting or advisory role for Merrimack (Inst) and Zeno Pharmaceuticals, and has received research funding from Aduro Biotech (Inst), Bristol-Myers Squibb (Inst), Genentech (Inst), Lilly (Inst), Merck (Inst), Newlink genetics (Inst), Pfizer (Inst), PRISM BioLab (Inst), and Sanofi (Inst). FJC has served in consulting or advisory role for AstraZeneca, and has received research funding from GRAIL.

Footnotes

Conflict of Interest Disclosure:

All other authors do not have any conflicts of interest to disclose.

References:

  • 1.National Cancer Institute: Surveillance E, and End Results Program. Cancer Stat Facts: Pancreatic Cancer. Available at: https://seer.cancer.gov/statfacts/html/pancreas.html (Last accessed: December 30,2019).
  • 2.Yadav S, Sharma P, Zakalik D. Comparison of Demographics, Tumor Characteristics, and Survival Between Pancreatic Adenocarcinomas and Pancreatic Neuroendocrine Tumors: A Population-based Study. Am J Clin Oncol. 2018;41(5):485–91. [DOI] [PubMed] [Google Scholar]
  • 3.Hu C, Hart SN, Polley EC, Gnanaolivu R, Shimelis H, Lee KY, et al. Association Between Inherited Germline Mutations in Cancer Predisposition Genes and Risk of Pancreatic Cancer. Jama. 2018;319(23):2401–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Matsubayashi H, Takaori K, Morizane C, Maguchi H, Mizuma M, Takahashi H, et al. Familial pancreatic cancer: Concept, management and issues. World journal of gastroenterology. 2017;23(6):935–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Stadler ZK, Salo-Mullen E, Patil SM, Pietanza MC, Vijai J, Saloustros E, et al. Prevalence of BRCA1 and BRCA2 mutations in Ashkenazi Jewish families with breast and pancreatic cancer. Cancer. 2012;118(2):493–9. [DOI] [PubMed] [Google Scholar]
  • 6.Salo-Mullen EE, O’Reilly EM, Kelsen DP, Ashraf AM, Lowery MA, Yu KH, et al. Identification of germline genetic mutations in patients with pancreatic cancer. Cancer. 2015;121(24):4382–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518(7540):495–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cancer Genome Atlas Research Network. Integrated Genomic Characterization of Pancreatic Ductal Adenocarcinoma. Cancer cell. 2017;32(2):185–203.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lohse I, Borgida A, Cao P, Cheung M, Pintilie M, Bianco T, et al. BRCA1 and BRCA2 mutations sensitize to chemotherapy in patient-derived pancreatic cancer xenografts. British journal of cancer. 2015;113(3):425–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Porcelli L, Quatrale AE, Mantuano P, Leo MG, Silvestris N, Rolland JF, et al. Optimize radiochemotherapy in pancreatic cancer: PARP inhibitors a new therapeutic opportunity. Molecular oncology. 2013;7(3):308–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Teo MY, O’Reilly EM. Is it time to split strategies to treat homologous recombinant deficiency in pancreas cancer? Journal of gastrointestinal oncology. 2016;7(5):738–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. The New England journal of medicine. 2019;381(4):317–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Golan T, Sella T, O’Reilly EM, Katz MH, Epelbaum R, Kelsen DP, et al. Overall survival and clinical characteristics of BRCA mutation carriers with stage I/II pancreatic cancer. British journal of cancer. 2017;116(6):697–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Blair AB, Groot VP, Gemenetzis G, Wei J, Cameron JL, Weiss MJ, et al. BRCA1/BRCA2 Germline Mutation Carriers and Sporadic Pancreatic Ductal Adenocarcinoma. Journal of the American College of Surgeons. 2018;226(4):630–7.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Yurgelun MB, Chittenden AB, Morales-Oyarvide V, Rubinson DA, Dunne RF, Kozak MM, et al. Germline cancer susceptibility gene variants, somatic second hits, and survival outcomes in patients with resected pancreatic cancer. Genetics in medicine : official journal of the American College of Medical Genetics. 2019;21(1):213–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zhu Y, Zhai K, Ke J, Li J, Gong Y, Yang Y, et al. BRCA1 missense polymorphisms are associated with poor prognosis of pancreatic cancer patients in a Chinese population. Oncotarget. 2017;8(22):36033–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Martin M Cutadapt Removes Adapter Sequences From High-Throughput Sequencing Reads [Available from: http://journal.embnet.org/index.php/embnetjournal/article/view/200.
  • 18.Li H Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM 2013. [November 29, 2018]. Available from: https://arxiv.org/abs/1303.3997. [Google Scholar]
  • 19.DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nature genetics. 2011;43(5):491–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wang C, Evans JM, Bhagwate AV, Prodduturi N, Sarangi V, Middha M, et al. PatternCNV: a versatile tool for detecting copy number changes from exome sequencing data. Bioinformatics (Oxford, England). 2014;30(18):2678–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kocher JP, Quest DJ, Duffy P, Meiners MA, Moore RM, Rider D, et al. The Biological Reference Repository (BioR): a rapid and flexible system for genomics annotation. Bioinformatics (Oxford, England). 2014;30(13):1920–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Liu X, Wu C, Li C, Boerwinkle E. dbNSFP v3.0: A One-Stop Database of Functional Predictions and Annotations for Human Nonsynonymous and Splice-Site SNVs. Human mutation. 2016;37(3):235–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Landrum MJ, Lee JM, Benson M, Brown G, Chao C, Chitipiralla S, et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic acids research. 2016;44(D1):D862–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Munz M, Ruark E, Renwick A, Ramsay E, Clarke M, Mahamdallie S, et al. CSN and CAVA: variant annotation tools for rapid, robust next-generation sequencing analysis in the clinical setting. Genome medicine. 2015;7:76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Khosrow-Khavar F, Filion KB, Al-Qurashi S, Torabi N, Bouganim N, Suissa S, et al. Cardiotoxicity of aromatase inhibitors and tamoxifen in postmenopausal women with breast cancer: a systematic review and meta-analysis of randomized controlled trials. Annals of oncology : official journal of the European Society for Medical Oncology. 2017;28(3):487–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hart SN, Duffy P, Quest DJ, Hossain A, Meiners MA, Kocher JP. VCF-Miner: GUI-based application for mining variants and annotations stored in VCF files. Briefings in bioinformatics. 2016;17(2):346–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genetics in medicine : official journal of the American College of Medical Genetics. 2015;17(5):405–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Krajewska M, Fehrmann RSN, de Vries EGE, van Vugt MATM. Regulators of homologous recombination repair as novel targets for cancer treatment. Frontiers in Genetics. 2015;6(96). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Golan T, Kanji ZS, Epelbaum R, Devaud N, Dagan E, Holter S, et al. Overall survival and clinical characteristics of pancreatic cancer in BRCA mutation carriers. British journal of cancer. 2014;111(6):1132–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lowery MA, Kelsen DP, Stadler ZK, Yu KH, Janjigian YY, Ludwig E, et al. An emerging entity: pancreatic adenocarcinoma associated with a known BRCA mutation: clinical descriptors, treatment implications, and future directions. The oncologist. 2011;16(10):1397–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Tran B, Moore S, Zogopoulos G, Borgida A, Holter S, Gallinger S, et al. Platinum-based chemotherapy (Pt-chemo) in pancreatic adenocarcinoma (PC) associated with BRCA mutations: A translational case series. Journal of Clinical Oncology. 2012;30(4_suppl):217-. [Google Scholar]
  • 32.Aung KL, Holter S, Borgida A, Connor A, Pintilie M, Dhani NC, et al. Overall survival of patients with pancreatic adenocarcinoma and BRCA1 or BRCA2 germline mutation. Journal of Clinical Oncology. 2016;34(15_suppl):4123-. [Google Scholar]
  • 33.Reiss KA, Yu S, Judy R, Symecko H, Nathanson KL, Domchek SM. Retrospective Survival Analysis of Patients With Advanced Pancreatic Ductal Adenocarcinoma and Germline BRCA or PALB2 Mutations. JCO Precision Oncology. 2018(2):1–9. [DOI] [PubMed] [Google Scholar]
  • 34.Dong X, Li Y, Hess KR, Abbruzzese JL, Li D. DNA mismatch repair gene polymorphisms affect survival in pancreatic cancer. The oncologist. 2011;16(1):61–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kondo T, Kanai M, Kou T, Sakuma T, Mochizuki H, Kamada M, et al. Association between homologous recombination repair gene mutations and response to oxaliplatin in pancreatic cancer. Oncotarget. 2018;9(28):19817–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sehdev A, Gbolahan O, Hancock BA, Stanley M, Shahda S, Wan J, et al. Germline and Somatic DNA Damage Repair Gene Mutations and Overall Survival in Metastatic Pancreatic Adenocarcinoma Patients Treated with FOLFIRINOX. Clinical cancer research : an official journal of the American Association for Cancer Research. 2018;24(24):6204–11. [DOI] [PubMed] [Google Scholar]
  • 37.Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491(7424):399–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science (New York, NY). 2008;321(5897):1801–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nature communications. 2015;6:6744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lowery MA, Wong W, Jordan EJ, Lee JW, Kemel Y, Vijai J, et al. Prospective Evaluation of Germline Alterations in Patients With Exocrine Pancreatic Neoplasms. JNCI: Journal of the National Cancer Institute. 2018;110(10):1067–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Park W, Chen J, Chou JF, Varghese AM, Yu KH, Wong W, et al. Genomic Methods Identify Homologous Recombination Deficiency in Pancreas Adenocarcinoma and Optimize Treatment Selection. Clinical cancer research : an official journal of the American Association for Cancer Research. 2020;26(13):3239–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Pishvaian MJ, Blais EM, Brody JR, Rahib L, Lyons E, Arbeloa PD, et al. Outcomes in Patients With Pancreatic Adenocarcinoma With Genetic Mutations in DNA Damage Response Pathways: Results From the Know Your Tumor Program. JCO Precision Oncology. 2019(3):1–10. [DOI] [PubMed] [Google Scholar]
  • 43.Yu S, Agarwal P, Mamtani R, Symecko H, Spielman K, O’Hara M, et al. Retrospective Survival Analysis of Patients With Resected Pancreatic Ductal Adenocarcinoma and a Germline BRCA or PALB2 Mutation. JCO Precision Oncology. 2019(3):1–11. [DOI] [PubMed] [Google Scholar]
  • 44.Kim H, Saka B, Knight S, Borges M, Childs E, Klein A, et al. Having Pancreatic Cancer with Tumoral Loss of ATM and Normal TP53 Protein Expression Is Associated with a Poorer Prognosis. Clinical Cancer Research. 2014;20(7):1865–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Weigelt B, Bi R, Kumar R, Blecua P, Mandelker DL, Geyer FC, et al. The Landscape of Somatic Genetic Alterations in Breast Cancers From ATM Germline Mutation Carriers. Journal of the National Cancer Institute. 2018;110(9):1030–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Nakamori S, Yashima K, Murakami Y, Ishikawa O, Ohigashi H, Imaoka S, et al. Association of p53 Gene Mutations with Short Survival in Pancreatic Adenocarcinoma. Japanese Journal of Cancer Research. 1995;86(2):174–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Xiang J-F, Wang W-Q, Liu L, Xu H-X, Wu C-T, Yang J-X, et al. Mutant p53 determines pancreatic cancer poor prognosis to pancreatectomy through upregulation of cavin-1 in patients with preoperative serum CA19–9 ≥ 1,000 U/mL. Scientific Reports. 2016;6(1):19222. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 48.Li VD, Li KH, Li JT. TP53 mutations as potential prognostic markers for specific cancers: analysis of data from The Cancer Genome Atlas and the International Agency for Research on Cancer TP53 Database. J Cancer Res Clin Oncol. 2019;145(3):625–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.NCCN Clinical Practice Guidelines in Oncology. Genetic/Familial High-Risk Assessment: Breast, Ovarian and Pancreatic Version 1.2020. : National Comprehensive Cancer Network; [12.08.2019]. Available from: https://www.nccn.org/professionals/physician_gls/pdf/genetics_bop.pdf. [Google Scholar]
  • 50.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. The New England journal of medicine. 2016;375(5):443–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Shroff RT, Hendifar A, McWilliams RR, Geva R, Epelbaum R, Rolfe L, et al. Rucaparib Monotherapy in Patients With Pancreatic Cancer and a Known Deleterious BRCA Mutation. JCO Precision Oncology. 2018(2):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Binder KAR, Mick R, O’Hara M, Teitelbaum U, Karasic T, Schneider C, et al. Abstract CT234: A Phase II, single arm study of maintenance rucaparib in patients with platinum-sensitive advanced pancreatic cancer and a pathogenic germline or somatic mutation in BRCA1, BRCA2 or PALB2. Cancer research. 2019;79(13 Supplement):CT234-CT. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES