Purpose:

Family history of BRCA-related tumors may correlate with response to chemotherapy and overall survival (OS) in pancreatic cancer. The frequency of germline mutations has been reported in patients predominantly under the age of 60 or with strong family history. We examine the incidence of deleterious germline mutations and compare the chemotherapy responses and OS in an unselected group of patients with metastatic pancreatic cancer.

Experimental Design:

Patients with metastatic pancreatic cancer, who were seen at a single cancer center between 2010 and 2016, were included. Germline DNA was sequenced using a 263-gene panel to identify novel mutations (N = 133 MD Anderson cohort, N = 127 TCGA cohort). Chemotherapy response and OS were determined by review of medical records.

Results:

Deleterious germline mutations were identified in 26 of 133 patients (19.5%). Patients with DNA damage repair (DDR) gene mutations (ATM, BRCA1/2, CDKN2A, CHEK2, ERCC4, PALB2, n = 15) had an improved OS as compared with patients without (16.8 vs. 9.1 months, P = 0.03). Conversely, patients with other deleterious mutations had a trend toward worse OS. However, survival in the latter group was longer (P = NS) in those mutants initially treated with gemcitabine/nab-paclitaxel. A family history of multiple breast, ovarian, and pancreatic cancers was associated with DDR gene mutations and better survival.

Conclusions:

We have identified novel germline mutations that are prognostic for survival in patients with pancreatic cancer. We observe improved survival in patients with DDR gene mutations and worsened survival in patients with deleterious mutations in non-DDR genes.

Translational Relevance

In this study, we examine the incidence of deleterious germline mutations and compare the chemotherapy responses and overall survival in an unselected group of patients with metastatic pancreatic cancer. Germline DNA was sequenced using a 263-gene panel to identify novel mutations. Deleterious germline mutations were identified in 26 of 133 patients (19.5%). Patients with DNA damage repair gene mutations (ATM, BRCA1/2, CDKN2A, CHEK2, ERCC4, PALB2) had a statistically significant improved overall survival as compared with those patients without (16.8 vs. 9.1 months, P = 0.03). We have also identified novel germline mutations that are prognostic for survival in pancreatic cancer patients. A family history of multiple breast, ovarian, and pancreatic cancers was associated with DDR gene mutations and better overall survival.

Familial pancreatic cancer (PC), defined as having two or more first-degree family members who have been diagnosed with pancreatic cancer, is thought to account for 5% to 10% of pancreatic adenocarcinoma (1). Previous studies of mutation prevalence in familial pancreatic cancer, however, have focused on a limited set of genes, mostly dedicated to those genes within the DNA damage repair (DDR) pathway (ATM, BRCA1, and BRCA2) or related to hereditary Lynch syndrome (MLH1, MSH2, MSH6, PMS2; refs. 2–7). The National Comprehensive Cancer Network (NCCN) currently endorses genetic counseling for all patients with pancreatic cancer (8). A provocative family history of cancer or Ashkenazi Jewish heritage may prompt some physicians to order further genetic testing. Recent studies have shown that pancreatic cancer family history is not predictive of germline mutations (7, 9), highlighting the value of broad sequencing in unselected groups. Specific genes are frequently tested on the basis of known cancer syndromes (10). The prevalence of mutations among patients with pancreatic cancer, who are unselected for specific risk factors such as age at diagnosis or family cancer history, is a current topic of interest amongst oncologists. Next-generation sequencing enables testing for both commonly described familial mutations, as well as rarely described variants.

We previously reported family history as a biomarker for survival in pancreatic cancer (11). We found that patients with a strong family history of BRCA-related cancers (three or more first- to third-generation relatives with breast, ovarian, or pancreas cancer) had an overall survival (OS) nearly double of those with no such family history. Family history, however, is a subjective measure and surrogate for the underlying disease biology leading to predisposition to pancreatic cancer in families.

In addition, despite evidence for a predictive benefit to platinum agents in familial PC, most practitioners use performance status as the main determinate for treatment regimen decisions. Patients with metastatic pancreatic cancer with poorer performance status (PS > 1) often receive treatment with gemcitabine plus nab-paclitaxel (gem/nab-paclitaxel), rather than the more difficult to tolerate platinum-based regimen 5-fluoruracil, irinotecan, and oxaliplatin (FOLFIRINOX; refs. 12, 13).

In this study, we assessed the prevalence of deleterious germline mutations using a 263-gene panel in a population of patients with pancreatic cancer, treated with current standard-of-care (SOC) chemotherapy, who presented to a large academic cancer institution. We also used a secondary cohort, the pancreatic cancer genome atlas (TCGA) research network germline data, to confirm the validity of some of our findings (14). Our goal was to identify additional underlying genomic alterations leading to familial pancreatic cancer in an unselected cohort including their link to family history, clinical implications, response to SOC treatment, and survival outcomes.

Patient selection

All patients with metastatic pancreatic adenocarcinoma who were seen at MD Anderson Cancer Center (MDACC, Houston, TX) between January 2010 and January 2016, and who received first-line SOC chemotherapy, were eligible for chart review. Cases were identified retrospectively. All patients had consented for DNA banking for clinical research. Samples were obtained from the MDACC pancreas cancer tissue bank and the Center for Translational and Public Health Genomics at Duncan Family Institute of MDACC and Patient History database Program. Clinical and pathologic information was abstracted from chart review from the MDACC electronic medical record including cancer stage, cancer histology, family history, and record of clinical genetic testing. Family history was defined as first- through third-degree (out to first cousins) relatives with breast, ovarian, or pancreatic cancer diagnosed at any age. The gastrointestinal pathologists in the MDACC Department of Pathology and Laboratory Medicine reviewed all pancreatic cancer tumors. All patients consented to DNA banking and clinical research. Of the 233 patients identified, blood samples for germline DNA testing were available from 133 cases including 95 and 38 patients treated with first-line FOLFIRINOX and gem/nab-paclitaxel, respectively. When available, outside germline mutation testing results were incorporated into our database. As specimens were used for research purpose alone, testing was not performed under Clinical Laboratory Improvement Amendments regulations. Results were not returned to the families of study participants or used to make clinical decisions. The study was conducted under the auspice of the MDACC Institutional Review Board. The study conformed to the Declaration of Helsinki. Informed written consent was obtained from each subject. A consort diagram is available in Fig. 1.

Figure 1.

Consortium diagram of cohort selection.

Figure 1.

Consortium diagram of cohort selection.

Close modal

Validation cohort

Deidentified germline genomic data was obtained from TCGA, pancreatic adenocarcinoma cohort. DNA sequencing data as well as a limited amount of clinical information was collected from all 127 patients.

Next-generation sequencing

Peripheral blood leukocytes were used to collect germline DNA. DNA was extracted using a QIAGEN DNA Extraction Kit (Qiagen Inc.). Targeted panel capture was performed on 500 ng of genomic DNA per sample based on KAPA library prep (KAPA Biosystems) using the T200.1 Solid Tumor Cancer gene panel (263 genes) and paired-end multiplex sequencing of samples was performed using the Illumina HiSeq 2500 sequencing platform (15). Average coverage was 450×.

Variant calling

Paired-end reads in FastQ format generated by the Illumina platform were aligned to the human reference genome (UCSC Genome Browser, hg19) using Burrows-Wheeler Aligner (16) by allowing three mismatches with 2 in the first 40 seeding regions and aligned reads processed using the GATK Best Practices of duplicate removal, indel realignment, and base recalibration (17). Germline single-nucleotide substitutions were detected using Platypus (18). The following filtering criteria were used: (i) total read coverage of the variant > = 20, (ii) the variants should be on-target, exonic, and nonsilent, (iii) a population frequency threshold of 1% was used to filter out common variants in the databases of dbSNP129 (PMID: 11237013), 1000 Genomes Project (PMID: 23128226), Exome Aggregation Consortium (Exac; PMID: 27535533), and ESP6500 (PMID: 23201682). Specifically, variants with a population frequency larger than 3% in any individual ethnicity group in Exac were also filtered out. A schema of variant calling and number of variants seen at each filtering step are provided in Fig. 2A.

Figure 2.

Variant classification pipeline (A) and number of variants at each level of variant filtering (B).

Figure 2.

Variant classification pipeline (A) and number of variants at each level of variant filtering (B).

Close modal

Variant classification

Gene variants deemed deleterious were considered mutations as were private, nonsilent variants not filtered out by the criteria shown in Fig. 2A. Those variants suspected as deleterious without evidence of validation in the literature were classified as variants of unknown significance (VUS). Variants were classified using American College of Medical Genetics and Genomics recommendations. Supporting data was obtained from literature review using linkage, biochemical, clinical, functional, and statistical research for specific alterations (19, 20). Variants were cross-referenced to Clinvar, HGMD, and UMDBRCA genomic data banks for further variant determination (21–23).

Statistical analysis

Patient clinical characteristics of categorical variables such as race and gender are reported given frequencies and percentages. Continuous data and sequencing results were summarized with descriptive statistics. The χ2 test and Fisher exact test were used to evaluate the association between two categorical variables. Wilcoxon rank sum test was used to compare the distributions of continuous variables between two different groups. Univariate and multivariable logistic regression models were used to evaluate the association between the outcomes of interest and patient demographic covariates. All tests are two-sided and P values < 0.05 are considered statistically significant. All analyses were conducted using SPSS version 24 (SPSS).

Study patient characteristics

We identified 233 patients that met our study cohort selection criteria as shown in Fig. 1. The median and mean ages at diagnosis of the entire cohort were 62 and 61 years (range, 36–84 years), respectively. 136 patients were male (58.4%) and all patients were metastatic at diagnosis. 155 patients had a baseline ECOG score of 0–1, 12 with 2, and in 15 patients, the score was unknown. 178 (76.4%) patients had liver metastases at diagnosis. Nineteen patients had secondary cancers (8.2%) that included breast (n = 3), colorectal cancer (n = 3), lung (n = 3), prostate (n = 3), bladder (n = 2), ovarian (n = 1), melanoma (n = 1), and various hematologic malignancies (n = 3), as well as other cancers. Four (1.7%) of these patients had multiple cancers including two patients with lung cancer, both heavy smokers, one with bladder cancer, and one with squamous cell carcinoma of the maxilla. Blood samples for germline DNA testing were available from 133 cases. The median and mean ages of the sequenced cohort at diagnosis were 61 and 60 years (range, 36–84 years), respectively. Sequenced patients were more likely to have received first-line FOLFIRINOX than gem/nab-paclitaxel (95 vs. 38 patients, P = 0.03). Otherwise, no other significant differences were observed between the two groups. A summary of these patients is presented in Table 1. A comparison of the sequenced versus unsequenced patients is found in Supplementary Table S1.

Table 1.

Patient clinical and pathologic characteristics of sequenced cohort.

CharacteristicNo. of patients sequenced cohort (n = 133)Percent (%)
Age at diagnosis of disease, years 
 Median 61  
 Range 36–84  
Gender 
 Male 78 58.6 
 Female 55 41.4 
ECOG PS 
 0–1 97 72.9 
 2+ 13 9.8 
 Unknown 23 17.3 
Ethnicity 
Caucasian 111 83.4 
Black 11 8.3 
 Hispanic 5.3 
 Asian 1.5 
 Other 1.5 
Formal genetic screening 
 Yes 18 13.5 
 No 115 86.5 
Ashkenazi Jewish 
 No 122 91.7 
 Yes 5.3 
 Unknown 
BRCA Relatives 
 0–1 114 85.7 
 2 13 9.8 
 3+ 4.5 
Second cancer 
 No 124 93.2 
 Yes 6.8 
 Median Baseline CA 19-9 940  
First-line chemotherapy 
 FOLFIRINOX 95 71.4 
 Gem/nab-paclitaxel 38 28.6 
Response to chemotherapy 
PR 37 27.8 
SD 49 36.9 
PD 33 24.8 
Not evaluable 14 10.5 
CharacteristicNo. of patients sequenced cohort (n = 133)Percent (%)
Age at diagnosis of disease, years 
 Median 61  
 Range 36–84  
Gender 
 Male 78 58.6 
 Female 55 41.4 
ECOG PS 
 0–1 97 72.9 
 2+ 13 9.8 
 Unknown 23 17.3 
Ethnicity 
Caucasian 111 83.4 
Black 11 8.3 
 Hispanic 5.3 
 Asian 1.5 
 Other 1.5 
Formal genetic screening 
 Yes 18 13.5 
 No 115 86.5 
Ashkenazi Jewish 
 No 122 91.7 
 Yes 5.3 
 Unknown 
BRCA Relatives 
 0–1 114 85.7 
 2 13 9.8 
 3+ 4.5 
Second cancer 
 No 124 93.2 
 Yes 6.8 
 Median Baseline CA 19-9 940  
First-line chemotherapy 
 FOLFIRINOX 95 71.4 
 Gem/nab-paclitaxel 38 28.6 
Response to chemotherapy 
PR 37 27.8 
SD 49 36.9 
PD 33 24.8 
Not evaluable 14 10.5 

Of the 133 sequenced patients, 29 deleterious mutations were found in 26 (19.6%) patients. Of those patients with a deleterious mutation, the median and mean ages were 56 and 55 years (range, 36–70 years), respectively. Seventy-eight (58.6%) patients were male, 111 (84%) were Caucasian, and 7 (5.3%) were of Ashkenazi Jewish descent. Of the 6 (4.5%) sequenced patients with a family history of 3 or more first- through third-degree BRCA-related relatives, 3 (50%) were found to have a deleterious mutation, and one of these patients had two inherited mutations. Conversely, of the 26 patients with a deleterious mutation and 2 (7.7%) patients had no family history of cancer. There were 21 patients with 0–1, 2 patients with 2, and 3 patients with 3 affected first-, second-, or third-degree relatives with a BRCA-associated cancer (breast, ovarian, or pancreas). Twenty-two of the 26 patients received first-line therapy with FOLFIRINOX. Clinical and tumor pathologic features for the patients with a deleterious mutation are provided in Table 2.

Table 2.

Summary of sequenced patient characteristics by deleterious mutation status.

CovariateLevelsDeleterious mutation = NoDeleterious mutation = YesP
Sex Male 63(58.9%) 15(57.7%) 0.91 
 Female 44(41.1%) 11(42.3%) — 
Age at diagnosis >60 57(53.3%) 10(38.5%) 0.18 
 ≤60 50(46.7%) 16(61.5%) — 
Age at Diagnosis >45 99(92.5%) 21(80.8%) 0.07 
 ≤45 8(7.5%) 5(19.2%) — 
ECOG 0,1 77(86.5%) 20(95.3%) 0.27 
 ≥2 12(13.5%) 1(4.7%) — 
First-line treatment 73(68.2%) 22(84.6%) 0.10 
 34(31.8%) 4(15.4%) — 
BRCA Group ≥3 104(97.2%) 23(88.5%) 0.05 
 3(2.8%) 3(11.5%) — 
Family history breast cancer 78(72.9%) 16(61.5%) 0.25 
 29(27.1%) 10(38.5%) — 
Family history ovarian cancer 102(95.3%) 25(96.2%) 0.86 
 5(4.7%) 1(3.8%) — 
Family history pancreatic cancer 94(87.9%) 20 (76.9%) 0.15 
 13(12.1%) 6 (23.1%) — 
Family history any cancer 25(23.4%) 5(19.2%) 0.96 
 82(76.6%) 21(80.8%) — 
Ashkenazi Jewish 99(96.1%) 23(88.5%) 0.12 
 4(3.9%) 3(11.5%) — 
Recurrent_VUS 58(54.2%) 22(84.6%) 0.005 
 49(45.8%) 4(15.4%) — 
Personal_Hx_Cancer 100(93.5%) 25(96.2%) 0.51 
 7(6.5%) 1(3.8%) — 
CovariateLevelsDeleterious mutation = NoDeleterious mutation = YesP
Sex Male 63(58.9%) 15(57.7%) 0.91 
 Female 44(41.1%) 11(42.3%) — 
Age at diagnosis >60 57(53.3%) 10(38.5%) 0.18 
 ≤60 50(46.7%) 16(61.5%) — 
Age at Diagnosis >45 99(92.5%) 21(80.8%) 0.07 
 ≤45 8(7.5%) 5(19.2%) — 
ECOG 0,1 77(86.5%) 20(95.3%) 0.27 
 ≥2 12(13.5%) 1(4.7%) — 
First-line treatment 73(68.2%) 22(84.6%) 0.10 
 34(31.8%) 4(15.4%) — 
BRCA Group ≥3 104(97.2%) 23(88.5%) 0.05 
 3(2.8%) 3(11.5%) — 
Family history breast cancer 78(72.9%) 16(61.5%) 0.25 
 29(27.1%) 10(38.5%) — 
Family history ovarian cancer 102(95.3%) 25(96.2%) 0.86 
 5(4.7%) 1(3.8%) — 
Family history pancreatic cancer 94(87.9%) 20 (76.9%) 0.15 
 13(12.1%) 6 (23.1%) — 
Family history any cancer 25(23.4%) 5(19.2%) 0.96 
 82(76.6%) 21(80.8%) — 
Ashkenazi Jewish 99(96.1%) 23(88.5%) 0.12 
 4(3.9%) 3(11.5%) — 
Recurrent_VUS 58(54.2%) 22(84.6%) 0.005 
 49(45.8%) 4(15.4%) — 
Personal_Hx_Cancer 100(93.5%) 25(96.2%) 0.51 
 7(6.5%) 1(3.8%) — 

Abbreviations: F, FOLFIRINOX; G, gemcitabine/nab-paclitaxel; N, no; Y, yes.

Frequency of deleterious mutations

Of all sequenced patients, 4 (3.0%) had a BRCA1 (n = 2) or BRCA2 (n = 2) germline mutation. In addition, 6 (4.5%) patients had deleterious mutations in other pancreatic cancer predisposition genes including ATM (n = 3), CDKN2A (n = 1), PALB2 (n = 1), and TP53 (n = 1). Thus, 10 (7.5%) patients had an inherited mutation in a known pancreatic cancer predisposition gene. Six (4.5%) patients had mutations in other DDR genes including ERCC4 (n = 2) and the checkpoint gene, CHEK2 (n = 4). Nine patients had other mutations in cancer-associated genes including TERT (n = 3), AR (n = 2), CYP2C19 (n = 2), NF1 (n = 1), and SDHD (n = 1). One IL7R mutation was identified in a male patient with a deleterious frameshift deletion in CHEK2, one patient had both a HNF1A mutation and a RET mutation, and one patient with a deleterious CDKN2A frameshift insertion had an IL7R mutation as well. In total, 18 (13.5%) patients had a mutation in a gene associated with cancer risk. No patients carried definitive, pathologic, deleterious mutations in POLE, STK11, or Lynch-related genes, although VUSs were found in these genes (Supplementary Table S2). Table 3 lists the 29 mutations identified, mutation effect, and associated patient characteristics.

Table 3.

MDACC cohort list of deleterious mutations and associated clinical characteristics.

Study IDGeneMutationMutation effectRecurrent in germline pancreatic TCGA cohortAge at Dx (years)Ashkenazi JewishPersonal cancer historyFamily history of cancerReferred to genetic counselorOS (days)
FamilyHx21 AR c.T170A Nonsynonymous SNV 70 None Endometrial (S) 297 
FamilyHx45 AR c.C2395G Nonsynonymous SNV 55 None Breast (M), colorectal (PA), gastric (MU), prostate (F, B) 229 
FamilyHx36 ATM c.1024_1027del Frameshift deletion 62 None Male breast (F) 77 
FamilyHx49 ATM c.5352delC Frameshift deletion 63 None Esophageal (S), lung (M) 694 
FamilyHx17 ATM c.4736dupA Frameshift insertion 42 None Colorectal (MGM, MU) 287 
FamilyHx24 BRCA1 c.2576delC Frameshift deletion 40 None Breast (M, MA x 2), endometrial (MGM), ovarian(M) 457 
FamilyHx107 BRCA1 c.6749delC Frameshift deletion 36 None Glioblastoma (MC), pancreatic (B) 1320 
FamilyHx108 BRCA2 c.5578delAA Frameshift deletion 53 None Breast (S), gastric (PA), head and neck (B), melanoma (F, B), ovarian (PA x 2), prostate (F) 540 
FamilyHx109 BRCA2 * Frameshift deletion 43 None Breast (S), lung (MU) 1307 
FamilyHx41 CHEK2 c.C254T Nonsynonymous SNV 67 None Breast (S), leukemia (F, S x 2), unknown gynecologic (S) 96 
FamilyHx20 CHEK2 c.C1412T Nonsynonymous SNV 50 None None 96 
FamilyHx3217 CHEK2 c.T599C Nonsynonymous SNV 62 None Glioblastoma (PGC), leukemia (PGU), lung (F), pancreatic (PU), prostate (PGF), unknown(PGA) 925 
FamilyHx12 CHEK2 c.1229delC Frameshift deletion 50 None Hepatocellular carcinoma (MA), lung (MA), pancreatic (M) 253 
FamilyHx3413 CDKN2A c.131insAA Frameshift insertion 58 None Bladder (PC), breast (PA), colorectal (MC), melanoma (PA, PC x 2), pancreatic (M, B), prostate (MU), ovarian (MC) 723 
FamilyHx27 CYP2C19 c.A1G Nonsynonymous SNV 65 None Lung (M) 44 
FamilyHx3198 CYP2C19 c.A1G Nonsynonymous SNV 56 None Hepatocellular carcinoma (PU), Prostate (F) 922 
FamilyHx42 ERCC4 c.C2395T Nonsynonymous SNV 64 None Lung (PGF) 88 
FamilyHx3460 ERCC4 c.C2395T Nonsynonymous SNV 65 None Colorectal (MGM), Unknown type skin (PGF) 497 
FamilyHx95 HNF1A c.G92A Nonsynonymous SNV 59 Prostate Bladder (F), colorectal, lung (F, PGF, PC), pancreatic (PA) 424 
FamilyHx12 IL7R c.G617A Nonsynonymous SNV 50 None Hepatocellular carcinoma (MA), lung (MA), pancreatic (M) 253 
FamilyHx3413 IL7R c.G214C Nonsynonymous SNV 58 None Bladder (PC), breast (PA), colorectal (MC), melanoma (PA, PC x 2), pancreatic (M, B), prostate (MU), ovarian (MC) 723 
FamilyHx3728 NF1 c.T2C Nonsynonymous SNV 48 None Breast (S, PA), prostate (F, PU) 192 
Family Hx110 PALB2 c.G3A Nonsynonymous SNV 58 None Breast (PC), colorectal (MU x 2, PA), kidney (MU, PC), lung (F, B, PU, PGF, PC), head and neck (B), pancreatic (PA), prostate (MU), unknown (MC, PGM) 737 
FamilyHx95 RET c.A2372T Nonsynonymous SNV 59 None Bladder (F), colorectal, lung (F, PGF, PC), pancreatic (PA) 424 
FamilyHx33 SDHD c.C33A Stopgain 50 None Carney's triad: paraganglioma, chondrosarcoma, and GIST (M), pheochromocytoma (M), urothelial (M), prostate (F) 350 
FamilyHx59 TERT c.C1234T Nonsynonymous SNV 68 None None 37 
Family Hx88 TERT c.C1234T Nonsynonymous SNV 62 None Colorectal (F) 224 
FamilyHx1 TERT c.G2371A Nonsynonymous SNV 50 None Endometrial (M), osteosarcoma (F), thyroid (M) 178 
FamilyHx2 TP53 c.G229T Stopgain 39 Bilateral breast cancer Breast (M), melanoma (F), neuroendocrine tumor (PC), sarcoma (S), unknown (MGF, MU) 268 
Study IDGeneMutationMutation effectRecurrent in germline pancreatic TCGA cohortAge at Dx (years)Ashkenazi JewishPersonal cancer historyFamily history of cancerReferred to genetic counselorOS (days)
FamilyHx21 AR c.T170A Nonsynonymous SNV 70 None Endometrial (S) 297 
FamilyHx45 AR c.C2395G Nonsynonymous SNV 55 None Breast (M), colorectal (PA), gastric (MU), prostate (F, B) 229 
FamilyHx36 ATM c.1024_1027del Frameshift deletion 62 None Male breast (F) 77 
FamilyHx49 ATM c.5352delC Frameshift deletion 63 None Esophageal (S), lung (M) 694 
FamilyHx17 ATM c.4736dupA Frameshift insertion 42 None Colorectal (MGM, MU) 287 
FamilyHx24 BRCA1 c.2576delC Frameshift deletion 40 None Breast (M, MA x 2), endometrial (MGM), ovarian(M) 457 
FamilyHx107 BRCA1 c.6749delC Frameshift deletion 36 None Glioblastoma (MC), pancreatic (B) 1320 
FamilyHx108 BRCA2 c.5578delAA Frameshift deletion 53 None Breast (S), gastric (PA), head and neck (B), melanoma (F, B), ovarian (PA x 2), prostate (F) 540 
FamilyHx109 BRCA2 * Frameshift deletion 43 None Breast (S), lung (MU) 1307 
FamilyHx41 CHEK2 c.C254T Nonsynonymous SNV 67 None Breast (S), leukemia (F, S x 2), unknown gynecologic (S) 96 
FamilyHx20 CHEK2 c.C1412T Nonsynonymous SNV 50 None None 96 
FamilyHx3217 CHEK2 c.T599C Nonsynonymous SNV 62 None Glioblastoma (PGC), leukemia (PGU), lung (F), pancreatic (PU), prostate (PGF), unknown(PGA) 925 
FamilyHx12 CHEK2 c.1229delC Frameshift deletion 50 None Hepatocellular carcinoma (MA), lung (MA), pancreatic (M) 253 
FamilyHx3413 CDKN2A c.131insAA Frameshift insertion 58 None Bladder (PC), breast (PA), colorectal (MC), melanoma (PA, PC x 2), pancreatic (M, B), prostate (MU), ovarian (MC) 723 
FamilyHx27 CYP2C19 c.A1G Nonsynonymous SNV 65 None Lung (M) 44 
FamilyHx3198 CYP2C19 c.A1G Nonsynonymous SNV 56 None Hepatocellular carcinoma (PU), Prostate (F) 922 
FamilyHx42 ERCC4 c.C2395T Nonsynonymous SNV 64 None Lung (PGF) 88 
FamilyHx3460 ERCC4 c.C2395T Nonsynonymous SNV 65 None Colorectal (MGM), Unknown type skin (PGF) 497 
FamilyHx95 HNF1A c.G92A Nonsynonymous SNV 59 Prostate Bladder (F), colorectal, lung (F, PGF, PC), pancreatic (PA) 424 
FamilyHx12 IL7R c.G617A Nonsynonymous SNV 50 None Hepatocellular carcinoma (MA), lung (MA), pancreatic (M) 253 
FamilyHx3413 IL7R c.G214C Nonsynonymous SNV 58 None Bladder (PC), breast (PA), colorectal (MC), melanoma (PA, PC x 2), pancreatic (M, B), prostate (MU), ovarian (MC) 723 
FamilyHx3728 NF1 c.T2C Nonsynonymous SNV 48 None Breast (S, PA), prostate (F, PU) 192 
Family Hx110 PALB2 c.G3A Nonsynonymous SNV 58 None Breast (PC), colorectal (MU x 2, PA), kidney (MU, PC), lung (F, B, PU, PGF, PC), head and neck (B), pancreatic (PA), prostate (MU), unknown (MC, PGM) 737 
FamilyHx95 RET c.A2372T Nonsynonymous SNV 59 None Bladder (F), colorectal, lung (F, PGF, PC), pancreatic (PA) 424 
FamilyHx33 SDHD c.C33A Stopgain 50 None Carney's triad: paraganglioma, chondrosarcoma, and GIST (M), pheochromocytoma (M), urothelial (M), prostate (F) 350 
FamilyHx59 TERT c.C1234T Nonsynonymous SNV 68 None None 37 
Family Hx88 TERT c.C1234T Nonsynonymous SNV 62 None Colorectal (F) 224 
FamilyHx1 TERT c.G2371A Nonsynonymous SNV 50 None Endometrial (M), osteosarcoma (F), thyroid (M) 178 
FamilyHx2 TP53 c.G229T Stopgain 39 Bilateral breast cancer Breast (M), melanoma (F), neuroendocrine tumor (PC), sarcoma (S), unknown (MGF, MU) 268 

Abbreviations: B, brother; F, father; M, mother; MA, maternal aunt; MC, maternal cousin; MGF, maternal grandfather; MGM, maternal grandmother; MU, maternal uncle; PA, paternal aunt; PC, paternal cousin; PGA, paternal grand aunt; PGF, paternal grandfather; PGM, paternal grandmother; PGU, paternal grand uncle; PGC, paternal grand cousin; PU, paternal uncle; S, sister.

*Denotes patient with clinically reported BRCA2 mutation; patient was uncertain of the specific mutation.

VUSs

We identified at least one VUS in 123 (93.2%) patients, with as many as 10 variants found per patient. On average, patients had 2.6 variants per patient (Fig. 2B). The most commonly seen VUSs were in FANCA [n = 8, (6%)], NOTCH1 [n = 8, (6%)], ERCC4 [n = 7, (5%)], and FGFR4 [n = 7, (5%)], respectively. Seven patients had VUSs in known pancreatic cancer predisposition genes including 2 (1.5%) patients with PALB2 variants, 2 (1.5%) patients with PMS2 variants, and 3 (2.3%) patients with POLE variants, including two recurrent variants. All VUSs identified are listed in Supplementary Table S2.

Overall survival and response rates

The median OS from date of diagnosis of the 133 sequenced patients was 10.0 months [95% confidence interval (CI): 8.5–11.5 months]. Of the sequenced patients with a family history of 0–1, 2, or 3 or more first-, second-, or third-degree relatives with a BRCA-associated cancer (breast, ovarian, or pancreas), the median OS was 9.6, 8.4, and 23.7 months, respectively (P = 0.13, Fig. 3A). The median OS in patients with versus without a deleterious mutation was 10.2 versus 9.9 months (P = 0.25, Fig. 3B). The median OS of the 10 patients with a deleterious DDR (ATM, BRCA1, BRCA2, ERCC4, PALB2) gene mutation versus other sequenced patients was significantly longer than patients without a DDR mutation (17.9 vs. 9.6 months, P = 0.03, Fig. 3C). There was no difference in survival for those patients who were 60 years old or less compared with those over 60 (10.0 vs. 9.4 months, P = 0.91, data not shown). The median OS from the date of the start of chemotherapy was 9.6 versus 8.9 months (P = 0.47, Fig. 3D), in those sequenced patients who were treated with FOLFIRINOX versus gem/nab-paclitaxel, respectively.

Figure 3.

Overall survival of sequenced cohort stratified by the number of family members with BRCA-related tumors (A), overall survival of sequenced cohort stratified by deleterious mutation (B), overall survival of sequenced cohort stratified by DDR or DDR cell-cycle checkpoint mutation (C), and overall survival of sequenced cohort stratified by treatment (D).

Figure 3.

Overall survival of sequenced cohort stratified by the number of family members with BRCA-related tumors (A), overall survival of sequenced cohort stratified by deleterious mutation (B), overall survival of sequenced cohort stratified by DDR or DDR cell-cycle checkpoint mutation (C), and overall survival of sequenced cohort stratified by treatment (D).

Close modal

On univariate analysis, ECOG PS > 1, family history of pancreatic cancer, family history of any cancer, and presence of 3 or more affected family members with a BRCA-associated cancer, were all significant determinants of survival (Supplementary Table S3). However, on multivariate analysis, ECOG PS >2 (HR 2.37; 95% CI, 1.28–4.37; P = 0.006) and a history of 3 or more family members with BRCA-associated cancers remained significant (HR 0.55; 95% CI, 0.32–0.93, P = 0.03; Table 4). Univariate analysis of all 233 patients is provided in Supplementary Table S4; however, the only significant predicting factor on multivariate analysis was ECOG PS (data not shown).

Table 4.

Multivariate analysis of determinants of overall survival.

CovariateHR (95% CI)P
ECOG ≥ 2 (vs. 0, 1) 2.37 (1.28–4.37) 0.006 
Family history pancreatic cancer = Yes (vs. No) 0.55 (0.32–0.93) 0.03 
CovariateHR (95% CI)P
ECOG ≥ 2 (vs. 0, 1) 2.37 (1.28–4.37) 0.006 
Family history pancreatic cancer = Yes (vs. No) 0.55 (0.32–0.93) 0.03 

Of the patients who underwent sequencing, there were 0 complete responses (CR), 37 partial responses (PR), 49 patients with stable disease (SD), 33 with progressive disease (PD), and 14 that were not evaluable due to lack of follow up imaging or other reasons (Table 1). The overall response rate (ORR) was 27.8% (37 of 133 patients). The ORR of patients without deleterious mutations was 29% (31 of 107 patients). Of the 26 patients with deleterious mutations, 22 were treated with FOLFIRINOX and 4 patients were treated with gem/nab-paclitaxel (Supplementary Table S5). There were 0 CRs, 6 PRs, 8 SDs, 8 PDs, and 4 were not evaluable with an ORR of 23.1% (6 of 26 patients). However, of the patients with DDR mutations, there were 0 CRs, 5 PRs, 6 SDs, 3 PDs, and 1 was not evaluable (ORR = 33.3%, 5 of 15 patients) compared with 0 CRs, 1 PR, 1 SD, 4 PD, and 2 who were not evaluable in those without DDR mutations (ORR = 12.5%).

Predictors of deleterious mutations

The prevalence of deleterious mutations decreased with increasing age at pancreatic cancer diagnosis, with frequencies of 41.7% (5/12), 20.4% (11/54), and 14.9% (10/67) for age groups younger than 45 years, 45 to 60 years, and older than 60 years, respectively. In a univariate analysis, age less than 45 nearly reached statistical significance (OR 2.94; 95% CI, 0.88–9.91; P = 0.08; Supplementary Table S6). Nearly half of all sequenced patients (56/133) had a family history of a BRCA-related cancer. There was a trend among those patients with three or more relatives with a BRCA-related cancer to have a deleterious mutation; however, this did not reach the level of statistical significance (OR = 4.53; 95% CI, 0.86–23.85; P = 0.08; Supplementary Table S5). Of all sequenced patients, 6.8% (9/133) had a personal history of an additional malignancy, including 3 patients with breast cancers. Personal history, however, did not predict for deleterious mutations (P = 0.96, Supplementary Table S6). There was a trend toward increased number of mutations in those with Ashkenazi Jewish heritage, although this did not reach statistical significance on univariate analysis. Other correlates are presented in Supplementary Table S5.

On multivariate analysis, only recurrent VUS was associated with a deleterious mutation (OR = 0.20; 95% CI, 0.06–0.62; P = 0.005; Supplementary Table S7).

The Cancer Genome Atlas validation

We additionally performed a similar query with the limited clinical data and germline sequencing data from the pancreatic cancer patients in TCGA. Median and mean age at diagnosis for this cohort was 61 (35–88 range, years) and sixty-five (51.2%) were male. Using the same variant calling pipeline (Fig. 2A), we identified 26 deleterious mutations in 24 (18.9%) patients of the 127 patients tested. There were no BRCA1 mutations and 2 (1.6%) BRCA2 mutations. Five (3.9%) patients had mutations in ATM, three (2.4%) patients had mutations in MUTYH, two patients each (1.6%) had mutations in AR, CHEK2, DNMT3A and one (0.8%) patient each had a mutation in CDKNA, CYP2C19, ERCC2, FANCA, IL7R, MEN1, NBN, PALB2, RET and TERT. Recurrent deleterious mutations that overlapped with our own study cohort were seen in 6 (4.7%) patients (Table 3). The median OS from date of diagnosis was 19.8 months in this cohort. We see that the median survival of patients with a DDR mutation exceeds that of those without DDR mutations (24.2 vs. 19.7 months, P = 0.024, data not shown).

Our current understanding of the prevalence of familial pancreatic cancer is largely limited to twelve genes previously described by Grant and colleagues in the Journal of Gastroenterology in 2015 (2). The incidence of damaging mutations in these genes, including ATM, BRCA1, BRCA2, and TP53 is thought to occur in 5% to 10% of patients with pancreatic cancer and is mainly limited to particular genes in the DDR pathways. In this study, we have evaluated for a larger variety of mutations that may predispose to familial pancreatic cancer and found a mutation rate of almost 20% when testing 263 cancer-associated genes in this unselected cohort.

Here, we have also provided additional evidence that family history of 3 or more BRCA-related cancers is a determinate of OS; however, this was not statistically significant. The small number of patients in this group and the subjective nature of family history may contribute to the lack of significance. Interestingly, patients with mutations in genes involved in DDR or the cell-cycle checkpoint (ATM, BRCA1, BRCA2, CDKN2A, CHEK2, PALB2, and ERCC4) had a near doubling of the OS (17.9 vs. 9.6 months, P = 0.03) compared with those that did not have a mutation in one of these genes. Perhaps not all deleterious mutations are beneficial. As seen in Table 3, there is a trend toward better outcomes in those with BRCA mutations than those with ERCC4.

Familial pancreatic cancer due to mutations in mismatch repair genes may demonstrate overlapping tumor biology with those with DNA repair defects. We did not find any pathologic mutations in MMR genes other than 2 VUSs in PMS2. In previous studies of familial pancreatic cancer, the estimated incidence of MMR gene mutations was very low at 0.1%–1% (24, 25). With a larger study cohort, we may have observed mutations in these genes. Future study may also include a broader range of genes related to DNA repair such as MREIIA, NBN, and BARD1.

The frequency of each DDR mutation in the TCGA cohort differs from our own discovery cohort. This is consistent with findings from Johns Hopkins University (Baltimore, MD) that showed a large amount of heterogeneity among patients with familial pancreatic cancer (26). However, when taken as a whole, the TCGA patients with DDR mutations also demonstrated prolonged OS (numbers), confirming our own findings. In 2019, a group at Memorial Sloan Kettering Cancer Center published a study in the journal, Genetics in Medicine, where they found a 10% incidence of germline pathogenic mutations in a cohort of patients with familial pancreatic cancer (25). They tested a larger cohort of patients, but used a smaller panel of genes to test both germline and somatic mutations. Importantly, we confirm that patients with pathogenic germline defects in DNA repair genes have a better overall survival. This may argue for a biologically based survival benefit in these patients either due to improved treatment response or less aggressive tumor biology. In the future, we plan to compare germline and somatic variants in our patient cohort.

In the clinic, decisions whether to treat patients with either of the current SOC regimens, FOLFIRINOX or gem/nab-paclitaxel, is largely based on performance status. Those in better condition tend to receive the more difficult to tolerate, yet arguably more effective, FOLFIRINOX. We did find that the majority of patients identified as carrying deleterious mutations in predisposition genes were treated with first-line FOLFIRINOX (85%). Of the 4 patients with mutations that received first-line gemcitabine and nab-paclitaxel, only two were evaluable for response. This may be due to the fact that FOLFIRINOX had become SOC by 2011 for first-line metastatic pancreatic cancer and it was not until 2013 that gem/nab-paclitaxel became FDA approved. In those with BRCA-mutated cancers, previous predictive data has largely been based on treatment with cisplatin, a chemotherapeutic agent that is less frequently used with current SOC chemotherapy regimens (27). Because of this widely held perspective, patients with a stronger family history, which may be more likely to represent those with a deleterious mutation, were placed on FOLFIRINOX. If patients with deleterious mutations in DDR genes have a better prognosis, they may be in better physical condition at presentation, and given the more difficult-to-tolerate regimen by their oncologist. Unfortunately, we did not have enough patients to characterize the effectiveness of these regimens among patients with inherited mutations.

One of the limitations of this study is that the vast majority of sequenced patients were deceased. This introduces a retrospective bias, as patients who lived longer might not undergo genetic testing. Therefore, the actual overall survival of mutated patients may be longer than what we report in this study. We would ideally perform a prospective study looking at an unselected pancreatic cancer cohort where all-comers are sequenced for germline mutations. In addition, mutations, although predicted to be deleterious, may not necessarily be causative of pancreatic adenocarcinoma. Furthermore, in this study, we focused on metastatic patients. We therefore, cannot speak to the germline mutation spectrum of resectable or locally advanced patients.

Only 13.5% of sequenced patients underwent formal genetic screening. Of those that underwent screening, mutations were found in 50% (9/18) of these patients. This was due to lack of follow through in sending blood for sequencing or a determination that sequencing was not necessary. We additionally compared our profiling with commercial tests. Two commercial labs, which omit testing for ERCC4 and CHEK2, would have missed 6 patients with DDR mutant cancers. The age of these affected patients ranged from 50 to 66. As these mutations occurred in patients over the age of 45, their omission would have potentially failed to predict cancers in relatives. The prevalence of ERCC4 and CHEK2 mutations among patients 50 and older suggests that commercial platforms should be expanded to include these genes. This also demonstrates the need for a more universal approach to evaluating familial pancreatic cancer.

In conclusion, we have better characterized the relationship of moderate penetrance germline mutation to familial pancreatic cancer. We have shown that deleterious mutations in either DDR genes or cell-cycle checkpoint genes confer a better prognosis over those without these mutations. Available commercial testing omits genes that may confer prognostic information. In addition, this study highlights the need for continued family counseling, preventative imaging, and early detection in those unaffected carriers. Further prospective studies can address these critical questions.

M.M. Javle is a consultant/advisory board member for Taiho, QED, Merck, and EMD Serono. No potential conflicts of interest were disclosed by the other authors.

Conception and design: J.B. Goldstein, M.M. Javle, F. McAllister, D.R. Fogelman

Development of methodology: J.B. Goldstein, M.M. Javle, D.R. Fogelman

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J.B. Goldstein, Y. Ghelman, M.M. Javle, R.T. Shroff, G.R. Varadhachary, R.A. Wolf, D.R. Fogelman

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J.B. Goldstein, L. Zhao, X. Wang, M.J. Overman, F. McAllister, A. Futreal, D.R. Fogelman

Writing, review, and/or revision of the manuscript: J.B. Goldstein, L. Zhao, X. Wang, M.J. Overman, R.T. Shroff, F. McAllister, A. Futreal, D.R. Fogelman

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): J.B. Goldstein, Y. Ghelman

The authors thank MDACC pancreas cancer tissue bank and the Center for Translational and Public Health Genomics at Duncan Family Institute of MDACC and Patient History database Program. This work was supported in part by the Welch Foundation's Robert A. Welch Distinguished University Chair Award (G-0040).

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.
Brand
RE
,
Lerch
MM
,
Rubinstein
WS
,
Neoptolemos
JP
,
Whitcomb
DC
,
Hruban
RH
, et al
Advances in counselling and surveillance of patients at risk for pancreatic cancer
.
Gut
2007
;
56
:
1460
9
.
2.
Grant
RC
,
Selander
I
,
Connor
AA
,
Selvarajah
S
,
Borgida
A
,
Briollais
L
, et al
Prevalence of germline mutations in cancer predisposition genes in patients with pancreatic cancer
.
Gastroenterology
2015
;
148
:
556
64
.
3.
Kastrinos
F
,
Mukherjee
B
,
Tayob
N
,
Wang
F
,
Sparr
J
,
Raymond
VM
, et al
Risk of pancreatic cancer in families with Lynch syndrome
.
JAMA
2009
;
302
:
1790
5
.
4.
Lin
KM
,
Shashidharan
M
,
Thorson
AG
,
Ternent
CA
,
Blatchford
GJ
,
Christensen
MA
, et al
Cumulative incidence of colorectal and extracolonic cancers in MLH1 and MSH2 mutation carriers of hereditary nonpolyposis colorectal cancer
.
J Gastrointest Surg
1998
;
2
:
67
71
.
5.
Moran
A
,
O'Hara
C
,
Khan
S
,
Nebgen
D
,
Peterson
SK
,
Singletary
C
, et al
Risk of cancer other than breast or ovarian in individuals with BRCA1 and BRCA2 mutations
.
Fam Cancer
2012
;
11
:
235
42
.
6.
Roberts
NJ
,
Jiao
Y
,
Yu
J
,
Kopelovich
L
,
Petersen
GM
,
Bondy
ML
, et al
ATM mutations in patients with hereditary pancreatic cancer
.
Cancer Discov
2012
;
2
:
41
6
.
7.
Shindo
K
,
Yu
J
,
Suenaga
M
,
Fesharakizadeh
S
,
Cho
C
,
Macgregor-Das
A
, et al
Deleterious germline mutations in patients with apparently sporadic pancreatic adenocarcinoma
.
J Clin Oncol
2017
;
35
:
3382
90
.
8.
National Comprehensive Cancer Network
. 
Genetic/familial high-risk assessment: breast and ovarian (Version 2.2019)
.
Available from
: https://www.nccn.org/professionals/physician_gls/pdf/genetics_screening.pdf; 
2018
.
9.
Bannon
SA
,
Montiel
MF
,
Goldstein
JB
,
Dong
W
,
Mork
ME
,
Borras
E
, et al
High prevalence of hereditary cancer syndromes and outcomes in adults with early-onset pancreatic cancer
.
Cancer Prev Res
2018
;
11
:
679
86
.
10.
Hampel
H
,
Bennett
RL
,
Buchanan
A
,
Pearlman
R
,
Wiesner
GL
;
Guideline Development Group
et al 
A practice guideline from the American College of Medical Genetics and Genomics and the National Society of Genetic Counselors: referral indications for cancer predisposition assessment
.
Genet Med
2015
;
17
:
70
87
.
11.
Fogelman
D
,
Sugar
EA
,
Oliver
G
,
Shah
N
,
Klein
A
,
Alewine
C
, et al
Family history as a marker of platinum sensitivity in pancreatic adenocarcinoma
.
Cancer Chemother Pharmacol
2015
;
76
:
489
98
.
12.
Conroy
T
,
Desseigne
F
,
Ychou
M
,
Bouché
O
,
Guimbaud
R
,
Bécouarn
Y
, et al
FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer
.
N Engl J Med
2011
;
364
:
1817
25
.
13.
Von Hoff
DD
,
Ervin
T
,
Arena
FP
,
Chiorean
EG
,
Infante
J
,
Moore
M
, et al
Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine
.
N Engl J Med
2013
;
369
:
1691
703
.
14.
The Cancer Genome Atlas
. 
The Cancer Genome Atlas - data portal
.
Available from
: tcga-data.nci.nih.gov.
15.
Chen
K
,
Meric-Bernstam
F
,
Zhao
H
,
Zhang
Q
,
Ezzeddine
N
,
Tang
LY
, et al
Clinical actionability enhanced through deep targeted sequencing of solid tumors
.
Clin Chem
2015
;
61
:
544
53
.
16.
Li
H
,
Durbin
R
. 
Fast and accurate short read alignment with Burrows-Wheeler transform
.
Bioinformatics
2009
;
25
:
1754
60
.
17.
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
.
Nat Genet
2011
;
43
:
491
8
.
18.
Rimmer
A
,
Phan
H
,
Mathieson
I
,
Iqbal
Z
,
Twigg
SRF
;
WGS500 Consortium
et al 
Integrating mapping-, assembly- and haplotype-based approaches for calling variants in clinical sequencing applications
.
Nat Genet
2014
;
46
:
912
18
.
19.
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
.
Genet Med
2015
;
17
:
405
24
.
20.
Eggington
JM
,
Bowles
KR
,
Moyes
K
,
Manley
S
,
Esterling
L
,
Sizemore
S
, et al
A comprehensive laboratory-based program for classification of variants of uncertain significance in hereditary cancer genes
.
Clin Genet
2014
;
86
:
229
37
.
21.
Stenson
PD
,
Ball
EV
,
Mort
M
,
Phillips
AD
,
Shiel
JA
,
Thomas
NS
, et al
Human Gene Mutation Database (HGMD): 2003 update
.
Hum Mutat
2003
;
21
:
577
81
.
22.
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 Res
2016
;
44
:
D862
8
.
23.
Beroud
C
,
Letovsky
SI
,
Braastad
CD
,
Caputo
SM
,
Beaudoux
O
,
Bignon
YJ
, et al
BRCA share: a collection of clinical BRCA gene variants
.
Hum Mutat
2016
;
37
:
1318
28
.
24.
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
:
2401
09
.
25.
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
.
Genet Med
2019
;
21
:
213
23
.
26.
Roberts
NJ
,
Norris
AL
,
Petersen
GM
,
Bondy
ML
,
Brand
R
,
Gallinger
S
, et al
Whole genome sequencing defines the genetic heterogeneity of familial pancreatic cancer
.
Cancer Discov
2016
;
6
:
166
75
.
27.
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
:
495
501
.