Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 May 1.
Published in final edited form as: Gastroenterology. 2019 Feb 1;156(6):1905–1913. doi: 10.1053/j.gastro.2019.01.254

Prevalence of Germline Mutations Associated with Cancer Risk in Patients With Intraductal Papillary Mucinous Neoplasms

Michael Skaro 1, Neha Nanda 1, Christian Gauthier 1, Matthäus Felsenstein 1, Zhengdong Jiang 1, Miaozhen Qiu 1,3, Koji Shindo 1, Jun Yu 2,4, Danielle Hutchings 1, Ammar A Javed 4, Ross Beckman 4, Jin He 4, Christopher L Wolfgang 1,2,4,5, Elizabeth Thompson 1,2, Ralph H Hruban 1,2,5, Alison P Klein 1,2,5,6, Michael Goggins 1,2,5,7, Laura D Wood 1,2,5, Nicholas J Roberts 1,2,5
PMCID: PMC6475492  NIHMSID: NIHMS1520497  PMID: 30716324

Abstract

Background & aims

Many patients with pancreatic adenocarcinoma (PDAC) carry germline mutations associated with increased risk of cancer. It is not clear whether patients with intraductal papillary mucinous neoplasms (IPMNs), which are precursors to some pancreatic cancers, also carry these mutations. We assessed the prevalence of germline mutations associated with cancer risk in patients with histologically confirmed IPMN.

Methods

We obtained non-tumor tissue from 315 patients with surgically resected IPMNs, from 1997 through 2017, and sequenced 94 genes with variants associated with cancer risk. Mutations associated with increased risk of cancer were identified and compared to individuals from the Exome Aggregation Consortium.

Results

We identified 23 patients with a germline mutation associated with cancer risk (7.3%; 95% CI, 4.9%–10.8%). Nine patients had a germline mutation associated with pancreatic cancer susceptibility (2.9% 95% CI, 1.4%–5.4%). More patients with IPMNs carried germline mutations in ATM (P<.0001), PTCH1 (P<.0001), and SUFU (P<.0001) compared with controls. Patients with IPMNs and germline mutations associated with pancreatic cancer were more like to have concurrent invasive pancreatic carcinoma compared to patients with IPMNs without these mutations (P<.0320).

Conclusions

In sequence analyses of 315 patients with surgically resected IPMNs, we found almost 3% to carry mutations associated with pancreatic cancer risk. More patients with IPMNs and germline mutations associated with pancreatic cancer had concurrent invasive pancreatic carcinoma compared to patients with IPMNs without these mutations. Genetic analysis of patients with IPMNs might identify those at greatest risk for cancer.

Keywords: Pancreas, cancer, genetics, predisposition

Introduction

Pancreatic adenocarcinoma (PDAC) is a deadly disease with a 5-year survival rate of just 8 percent1. By 2030, PDAC is predicted to become the second leading cause of cancer-related death in the United States1. Understanding the genetics and biology of pancreatic tumorigenesis is key to early diagnosis when patient outcomes are much improved2, 3. In particular, understanding the risk factors driving development of non-invasive pancreatic precursor lesions and their transition to invasive carcinoma is essential to appropriate patient stratification and intervention.

Approximately 10% of patients with PDAC have a germline mutation in an established pancreatic cancer susceptibility gene, including: ATM, BRCA1, BRCA2, CDKN2A, CPA1, MLH1, MSH2, PALB2, PMS2, PRSS1, and STK11412. Prevalence of a germline mutation is higher still in patients with PDAC and a family history of pancreatic cancer in a first-degree relative, reaching 15–20%4. Inheritance of a germline mutation in an established pancreatic cancer susceptibility gene can impact patient care in several ways. First, knowledge of germline status allows for informed, risk-appropriate screening strategies to be undertaken and PDAC to be detected early3, 13. Second, as many established susceptibility genes predispose to tumors in a number of organs, recommended screening for these extra-pancreatic cancers can be instituted14. Finally, in some patients with PDAC, germline mutation status may have therapeutic implications, for example, use of poly [ADP-ribose] polymerase-1 (PARP-1) inhibitors or platinum-based chemotherapy for tumors deficient in homology directed DNA due to BRCA2 loss and use of immunotherapy for patients with tumors deficient in mismatch repair due to loss of MLH1, MSH2, MSH6, or PMS21517.

PDAC forms when normal ductal epithelium acquires sequential genetic, cellular, and morphological alterations1821. These alterations are well-defined and result in progression from normal epithelium, to non-invasive precursor lesion, and finally invasive carcinoma22. Pre-malignant, non-invasive precursor lesions are of three types, microscopic pancreatic intraepithelial neoplasia and macroscopic intraductal papillary mucinous neoplasms (IPMNs) and mucinous cystic neoplasms23. As IPMNs are macroscopic and non-invasive, they represent an ideal opportunity for intervention before progression to PDAC. IPMNs, however, are common in the population24, 25 and numerous clinical criteria are used as surrogates of high-grade dysplasia or invasive cancer to identify IPMN patients with a high-risk of progression to PDAC and may benefit from surgical intervention. These include size of the main pancreatic duct, cyst size, presence of a mural nodule, and symptoms such as pancreatitis or jaundice2629. Although useful, these clinical criteria are imprecise and indirect measures of tumor biology. Molecular markers that indicate a need for surgical resection are desperately needed but are currently lacking.

Several lines of evidence suggest a possible underlying genetic predisposition to IPMNs. First, IPMNs are often multifocal and the remnant pancreas is at increased risk of IPMN after resection. This multifocality could be due intraluminal spread of neoplastic cells, to an environmental exposure, or an underlying genetic predisposition3032. Second, germline mutations in pancreatic cancer susceptibility genes such as BRCA2, CDKN2A, and STK11 have been identified in patients with IPMN3335. Third, in one screening study of 78 patients at high-risk of pancreatic cancer, most of the patients who underwent pancreatic resection for concerning imaging findings had IPMN36. And in another study, the prevalence of incipient and high-grade IPMN was higher in patients with familial compared to sporadic PDAC37. Finally, several reports have suggested that patients with an IPMN have an increased risk of developing other cancers, including colon cancer35, 3841.

Despite the potential ramifications of germline status in patients with IPMNs, no studies have systematically characterized germline mutations in this patient population. Therefore, we used targeted next-generation sequencing to characterize variation in genes that predispose to PDAC and other cancers in a series of 315 patients with surgically resected, histologically confirmed, IPMN.

Materials and methods

Patients and biospecimens

This study was reviewed and approved by the Johns Hopkins Medicine Institutional Review Board. 350 unselected patients with surgically resected IPMN and available non-tumor tissue were identified from surgical and pathology databases. Where available, 25 mg of fresh-frozen non-tumor tissue (duodenum) was obtained. Otherwise, 0.6 mm tissue cores were obtained from formalin-fixed blocks (FFPE) of non-tumor tissue (duodenum, gallbladder, liver, or spleen).

DNA extraction

DNA was extracted from fresh-frozen non-tumor tissue using the DNeasy Blood & Tissue Kit (Qiagen, catalog no. 69504) according to the manufacturer’s instructions. DNA from FFPE non-tumor tissue cores was extracted using the QIAamp DNA FFPE Tissue Kit (Qiagen, catalog no. 56404) and deparaffinization solution (Qiagen, catalog no. 19093) with the following protocol modifications: 1) 10 or fewer tissue cores were de-paraffinized with 120 μL of deparaffinization solution, while 11 or more tissue cores were deparaffinized with 200 μL of deparaffinization solution, 2) after addition of ATL buffer and proteinase K, samples were incubated for up to 7 days with intermittent mixing by inversion and vortex, and 3) an additional 20 μL of proteinase K was added to the sample after 48 hours of incubation. Extracted DNA was quantified with the Qubit 3.0 Fluorometer (Thermo Fisher Scientific) using the Qubit 1× dsDNA BR Assay Kit (Thermo Fisher Scientific, catalog no. Q32853).

Library preparation, sequencing, and analysis

DNA sequence libraries for each sample were prepared with the TruSight Rapid Capture Kit (Illumina, catalog no. FC-140–1105) and pooled into groups of 12 before capture with the TruSight Cancer probe set (Illumina, catalog no. FC-140–1101) according to the manufacturer’s instructions. The TruSight Cancer probe set covers the coding region of 94 hereditary cancer predisposition genes (Supplementary Table 1). Fragment size and yield of captured libraries were assessed with the Bioanalyzer 2100 Instrument (Agilent, catalog no. G2939BA) using the High Sensitivity DNA Kit (Agilent, catalog no. 5067–4626) and the Qubit 3.0 Fluorometer (Thermo Fisher Scientific) using the Qubit 1× dsDNA HS Assay Kit (Thermo Fisher Scientific, catalog no. Q33230). Captured sequence libraries were further pooled into groups of 24 samples and sequenced on the Illumina MiSeq System (Illumina, CA) using the MiSeq Reagent Kit v2 (300-cycles) (Illumina, catalog no. MS-102–2002), generating 150 base pair (bp) paired-end reads. Sequence reads were processed through a standardized pipeline using MiSeq Reporter Software v2.6 (Illumina, CA). Sequence reads were aligned to the human reference genome (hg19) using Burrows-Wheeler Aligner (BWA)42. Variant calling was performed with Genome Analysis Tool Kit (GATK)43. Samples with less than 20× average target coverage were excluded from analysis. Annotation of variants was conducted with ANNOVAR and included amino acid alterations based on RefSeq transcripts, minor allele frequency (MAF) using publicly available variant databases (1000 Genomes Project, Exome Variant Server, and Exome Aggregation Consortium (ExAC)), and ClinVar annotations4446. Variants (single base substitutions (SBS) or insertions/deletions (INDEL)) within exons or adjacent intronic sequence (+/−1, +/−2) of target genes were classified as either benign, of unknown significance, or deleterious germline mutation as follows: 1) benign – a variant of any functional consequence of > 0.5 % MAF or a synonymous variant of any MAF, 2) variant of unknown significance – a missense SBS or in-frame INDEL of ≤ 0.5 % MAF, and 3) deleterious – a frameshift or splicing INDEL, a nonsense SBS, a stop loss SBS, or splicing SBS of ≤ 0.5 % MAF. Sequence reads supporting deleterious germline variant calls were inspected using the Integrative Genomics Viewer47.

Variant validation

Putative deleterious germline mutations were validated via PCR amplification and Sanger sequencing of the variant region. Primers (Integrated DNA Technologies, Inc., CA) used for amplification are given in Supplementary Table 2. PCR set-up was conducted with OneTaq (NEB, catalog no. M0480S) according to manufacturer’s instructions. Amplification was conducted with the T100 Thermo Cycler (BioRad, catalog no. 1861096) using the following cycling conditions: one cycle of 94° C for 30 s, 21 cycles of 94° C for 30 s, 70° C for 30 s (decrement 0.5° C per cycle), 68° C for 60 s, and 25 cycles of 94° C for 30 s, 60° C for 30 s, 68° C for 60 s. PCR products were purified with the QIAquick PCR Purification Kit (Qiagen, catalog no. 28104) and Sanger sequenced (Genewiz, MD). Sequence chromatograms were visualized with 4Peaks (Nucleobytes, Netherlands)

Statistical analysis

Statistical analyses were conducted with Prism 6 (GraphPad Software). Confidence intervals for percent of samples with a hereditary cancer predisposition gene or pancreatic cancer susceptibility gene were calculated using the modified Wald method. Germline mutations in surgically resected IPMN patients and non-TCGA samples from ExAC were grouped by gene and compared using a two-tailed, chi-square test with Yates’ correction. Bonferroni correction for multiple testing was used and a P value < 5.3×10−4 was considered significant. Germline mutations in patients with surgically resected IPMN and unselected PDAC patients were grouped by gene and compared using a two-tailed Fisher’s exact test. Clinicopathologic variables in surgically resected IPMN patients by presence of germline mutation and invasive cancer were compared using a two-tailed Fisher’s exact test, except for age at time at surgery, duration of follow-up, and mean longest diameter of IPMN, which were compared using a two-tailed, unpaired t test. P values < 0.05 were considered significant. P values less than 0.0001 were abbreviated to < 0.0001.

Results

350 patients with surgically resected IPMN were included in this study. 315 patients had greater than 20× average target coverage after sequencing and were included in subsequent analyses. 138 patients had a high-grade IPMN (43.8%), 152 patients had a low- or intermediate-grade IPMN (48.3%), while 25 did not have a reported grade (7.9%). 62 (19.7%) patients had multifocal IPMN. 72 patients had IPMN and a co-occurring invasive carcinoma (22.9%), most commonly PDAC (57 patients). Other types of invasive carcinoma present in the study population included colloid carcinoma (11 patients), adenosquamous PDAC (1 patient), anaplastic carcinoma (1 patient), colloid carcinoma and PDAC (1 patient), and signet ring carcinoma (1 patient). 40 patients (12.7%) had a family history of pancreatic cancer in either a 1st or 2nd degree relative and 54 patients (17.1%) had a personal history of cancer. Further details of patient demographics and characteristics are given in Table 1 and Supplementary Table 3.

Table 1.

Demographics and characteristics of patients with surgically resected IPMN

Characteristic1 Number Percent
Race
White 270 85.7
Other 45 14.3
Sex
Male 162 51.4
Female 153 48.6
Age
<40 7 2.2
41–45 6 1.9
46–50 11 3.5
51–55 17 5.4
56–60 28 8.9
61–65 40 12.7
66–70 60 19.0
71–75 69 21.9
76–80 49 15.6
81–85 21 6.7
>86 7 2.2
Family history of pancreatic cancer
Yes 40 12.7
No 205 65.1
NR 70 22.2
Personal history of cancer
Yes 54 17.1
No 247 78.4
NR 14 4.4
Diagnosis
IPMN 243 77.1
IPMN and invasive carcinoma 72 22.9
Size of IPMN
<1 22 7.0
≥1 and <2 87 27.6
≥2 and <3 85 27.0
≥3 and <4 48 15.2
≥4 and <5 23 7.3
≥5 32 10.2
NR 18 5.7
Number of IPMN
1 253 80.3
2+ 62 19.7
Duct type
Branch duct 146 46.3
Main duct 112 35.6
NR 57 18.1
Grade of IPMN
High 138 43.8
Low or intermediate 152 48.3
NR 25 7.9
1

IPMN - intraductal papillary mucinous neoplasm. NR - not reported. Family history of pancreatic cancer in 1st and 2nd degree relatives.

Targeted sequencing generated a mean of 150 Mbp per sample (range: 10–562 Mbp; standard deviation: 138 Mbp). Mean target coverage was 256× (range: 20–877×; standard deviation: 140×). Mean target region covered at 1× and 10× was 99.1% (73.9–100%, standard deviation: 2.0%) and 97.2% (range: 46.9–100%; standard deviation: 5.6%) respectively. Mean number of SNVs identified per patient was 276 (range: 56–340; standard deviation: 40) and mean number of insertions and deletions was 1 (range: 1–3; standard deviation: 0).

Variants identified in the 94 hereditary cancer predisposition genes covered by the TruSight Cancer Panel were classified as either benign variant, variant of unknown significance, or deleterious germline mutations (see Materials and Methods). This analysis identified 26 germline mutations in 23 patients (7.3%: 95 percent confidence interval 4.9–10.8%) (Table 2). 10 germline mutations in 9 patients were in established pancreatic cancer susceptibility genes (2.9%: 95 percent confidence interval 1.3–5.4%), including five germline mutations in ATM, three germline mutations in BRCA2, one germline mutation in MSH6, and one germline mutation in PALB2. One germline mutation was also identified in BUB1B, a previously identified candidate pancreatic cancer susceptibility gene11. More than one patient had a germline mutation involving ATM (5 patients), BRCA2 (3 patients), FANCI (2 patients), and PTCH1 (2 patients). Three patients had more than one germline mutation in a hereditary cancer predisposition gene. One patient had both a RB1 and PTCH1 germline mutation, one patient had both a BRCA2 and FANCM germline mutation, and another had both a BRCA2 and MSH6 germline mutation. Similar findings have been reported for familial pancreatic cancer and familial pancreatitis in which affected individuals have deleterious germline mutations in multiple susceptibility genes11,48.

Table 2.

Germline mutations identified in patients with surgically resected IPMN

Patient number Gene Type Transcript Germline mutation1 Functional consequence Concurrent invasive carcinoma
1 ATM NM 000051 g.chr11:108098600 G>A c.G170A p.W57X Stopgain Signet ring carcinoma
2 ATM NM 000051 g.chr11: 108117812 CAAAG>C c.1024 1027del p.K342fs Frameshift deletion PDAC
3 ATM NM_000051 g.chr11:108137985_C>T c.C2554T p.Q852X Stopgain -
4 ATM Pancreatic NM 000051 g.chr11:108175549 C>T c.C5644T p.R1882X Stopgain PDAC
5 ATM cancer NM 000051 g.chr11:108206686 A>T c.A8266T p.K2756X Stopgain -
6 BRCA2 susceptibility NM 000059 g.chr13:32907014 A>T c.A1399T p.K467X Stopgain -
7 BRCA2 gene NM 000059 g.chr13:32914437 GT>G c.5946delT p.S1982fs Frameshift deletion -
8 BRCA2 NM 000059 g.chr13:32972346_TTGTA>T c.9697_9700del p.C3233fs Frameshift deletion Colloid carcinoma
6 MSH6 NA g.chr2:48033791 GTAAC>G - - Splicing -
9 PALB2 NM_024675 g.chr16:23649206_GACAA>G c.172_175del p.L58fs Frameshift deletion PDAC
10 ALK Hereditary NM 004304 g.chr2:29436851 G>A c.C3742T p.R1248X Stopgain -
11 BRIP1 cancer NM 032043 g.chr17:59871059 C>A c.G1372T p.E458X Stopgain Adenosquamous PDAC
12 BUB1B susceptibility NM 001211 g.chr15:40462282 C>T c.C199T p.R67X Stopgain PDAC
13 CDH1 gene NM 001317184 g.chr16:68771344 C>A c.C26A p.S9X Stopgain -
14 FANCA NA g.chr16:89871687 C>G - - Splicing -
15 FANCD2 NM 001018115 g.chr3:10083368 C>T c.C757T p.R253X Stopgain PDAC
16 FANCI NM 001113378 g.chr15:89838165 C>T c.C2476T p.Q826X Stopgain -
17 FANCI NM 018193 g.chr15:89843584 C>CA c.2678dupA p.Q893fs Frameshift insertion -
8 FANCM NM 001308133 g.chr14:45645855 G>T c.G3820T p.E1274X Stopgain Colloid carcinoma
18 NBN NM 002485 g.chr8:90960063 T>A c.A1903T p.K635X Stopgain -
19 PTCH1 NM 001083603 g.chr9:98279098 TC>T c.4delG p.E2fs Frameshift deletion Colloid carcinoma
20 PTCH1 NM 001083603 g.chr9:98279098 TC>T c.4delG p.E2fs Frameshift deletion -
20 RB1 NA g.chr13:48922000 G>A - - Splicing -
21 RECQL4 NM 004260 g.chr8:145739410 G>A c.C1960T p.Q654X Stopgain -
22 SUFU NM 001178133 g.chr10:104268965 CA>C c.223delA p.R75fs Frameshift deletion -
23 WT1 NM 000378 g.chr11:32456755 GC>G c.136delG p.A46fs Frameshift deletion -

IPMN - Intraductal papillary mucinous neoplasm, PDAC - pancreatic adenocarcinoma.

1

g - genomic change, c - transcript change; p - protein change associated with germline mutation. Genomic co-ordinates use hg19 version of human genome.

We next compared the prevalence of germline mutations in surgically resected IPMN patients to similarly-analyzed, publicly-available variant data from ExAC (Table 3)46. Germline mutations were not significantly enriched when considering all sequenced hereditary cancer predisposition genes (P value = 0.6590) or pancreatic cancer susceptibility genes (P value = 0.1403). Similarly, the majority of individual genes sequenced were not significantly enriched in patients with an IPMN. However, three genes were significantly enriched after Bonferroni correction for multiple testing. These genes are ATM (P value = < 0.0001), PTCH1 (P value = < 0.0001), and SUFU (P value = < 0.0001).

Table 3.

Comparison of germline mutations identified in patients with surgically resected IPMN and ExAC controls

Germline mutation IPMN EXAC
AC AN AF AC AN AF P value
Hereditary cancer gene 26 630 0.041 3921 105586 0.037 0.6590
Pancreatic cancer susceptibility gene 10 630 0.016 992 105732 0.009 0.1403
ATM 5 630 0.008 134 106203 0.001 <0.0001*
BRCA2 3 630 0.005 216 106188 0.002 0.2858
MSH6 1 630 0.002 261 106196 0.002 0.9709
PALB2 1 630 0.002 63 106206 0.001 0.8413
ALK 1 630 0.002 24 106209 0.000 0.3570
BRIP1 1 630 0.002 120 106202 0.001 0.7336
BUB1B 1 630 0.002 32 106209 0.000 0.4874
CDH1 1 630 0.002 9 96677 0.000 0.0861
FANCA 1 630 0.002 117 105585 0.001 0.7189
FANCD2 1 630 0.002 83 106209 0.001 0.9947
FANCI 2 630 0.003 83 106208 0.001 0.1569
FANCM 1 630 0.002 174 106183 0.002 0.9746
NBN 1 630 0.002 59 103676 0.001 0.7286
PTCH1 2 630 0.003 14 105834 0.000 <0.0001*
RB1 1 630 0.002 6 106198 0.000 0.0235
RECQL4 1 630 0.002 173 105674 0.002 0.9754
SUFU 1 630 0.002 0 105586 0.000 <0.0001*
WT1 1 630 0.002 13 105241 0.000 0.1476

IPMN - intraductal papillary mucinous neoplasm; ExAC - Exome Aggregation Consortium; AC - germline mutation allele count, AN - assessed allele number; AF - frequency of germline mutations.

*

Significant when applying Bonferroni correction for multiple testing (threshold for significance = 5.3×10−4).

We also compared the prevalence of germline mutations in established pancreatic cancer susceptibility genes between surgically resected IPMN patients and previously published series of unselected PDAC patients (Supplementary Table 4)8, 9. No genes analyzed had statistically significant over- or under-representation in surgically resected IPMN patients compared to unselected PDAC patients.

The patients with IPMN that had a germline mutation in a pancreatic cancer susceptibility gene were more likely to have concurrent invasive carcinoma than IPMN patients without a germline mutation. Specifically, 5 of 9 patients with germline mutation in a pancreatic cancer susceptibility gene had concurrent invasive carcinoma compared to 67 of 306 patients without a germline mutation (Fisher’s exact test; p-value = 0.0320) (Table 4). Interestingly, there was no statistically significant association between a germline mutation in a hereditary cancer predisposition gene and concurrent invasive carcinoma (Table 4). Of the five patients with a germline mutation in a pancreatic cancer susceptibility gene and invasive carcinoma, only one had a family history of pancreatic cancer in a 1st or 2nd degree relative and none had a reported previous cancer history. Otherwise, there were no statistically significant differences between IPMN patients with a germline line mutation in either a hereditary cancer predisposition gene or a pancreatic cancer susceptibility gene compared to IPMN patients without a germline mutation with respect to family history of pancreatic cancer in 1st or 2nd degree relatives, personal history of cancer, age at surgery, sex, presence of multifocal IPMN, high-grade dysplasia, size, or main duct involvement (Table 4).

Table 4.

Comparison of patients with surgically resected IPMN with and without a germline mutation

Variable1 Germline mutation in hereditary cancer predisposition gene Germline mutation in pancreatic cancer susceptibility gene
+
(n=23)

(n=292)
p-value +
(n=9)

(n=306)
P value
Patients with concurrent invasive carcinoma (n) 9 63 0.0694 5 67 0.0320
Patients with family history of pancreatic cancer (n) 6 34 0.0971 3 37 0.1670
Patients with personal history of cancer (n) 1 53 0.1419 1 53 1.0000
Mean age at surgery (years) 65.2 68.2 0.1911 62.2 68.2 0.1025
Male patients (n) 14 148 0.3916 6 156 0.5031
Patients with high-grade dysplasia (n) 8 130 0.6442 2 136 0.6865
Mean longest diameter of IPMN (cm) 2.1 2.7 0.0986 2.1 2.7 0.3674
Patients with multifocal IPMN (n) 4 58 1.0000 2 60 0.6921
Patients with main duct involvement (n) 6 106 1.0000 2 110 0.3078
Mean duration of follow-up (months) 46.8 32.5 0.1248 40.2 33.2 0.6287
Incident pancreatic cancer during follow-up (n) 0 2 1.0000 0 2 1.0000
1

Not all patients had a grade of dysplasia assigned. P-values calculated using samples with reported family history status (6/19, 34/226, 3/9, 37/236), reported personal cancer history (1/20, 53/281, 1/9, 53/292), grade assigned (8/19, 130/271, 2/6, and 136/284), main duct involvement (6/15, 105/243, 2/6, and 110/252), and incident pancreatic cancer during follow-up (0/15, 2/229, 0/6, and 2/238).

Patients with IPMN and invasive carcinoma were significantly more likely to have high-grade dysplasia (P value = < 0.0001) and involvement of the main pancreatic duct (P value = < 0.0059) compared to patients without concurrent invasive carcinoma (Supplementary Table 5). There were no other statistically significant associations between IPMN patients with and without invasive carcinoma.

Follow-up was available for 243 of 315 patients with a mean duration of 33.3 months (range: 0.1 – 199.3 months). The number of patients with a new diagnosis of pancreatic cancer during follow-up was 2 (0.8%). There were no significant differences in mean duration of follow-up or incident pancreatic cancers between patients with a germline mutation and those without a germline mutation (Table 4).

Discussion

In this retrospective study of patients with surgically resected, histologically confirmed, IPMN, we found that 7.3% of patients had a germline mutation in a hereditary cancer predisposition gene and 2.9% had a germline mutation in an established pancreatic cancer susceptibility gene. The number of patients with a germline mutation in a either a hereditary cancer predisposition gene or a pancreatic cancer susceptibility gene was not significant when compared to ExAC controls. However, prevalence of a germline mutation in pancreatic cancer susceptibility genes in IPMN patients is similar to recent studies of PDAC patients unselected for family history where between 3.9 and 5.5% patients had a germline mutation8, 9.

Three individual genes were significantly enriched in surgically resected IPMN patients compared to ExAC controls. These genes include ATM (five germline mutations), PTCH1 (two germline mutations), and SUFU (one germline mutation). ATM is a serine/threonine kinase integral to DNA double strand break repair in response to ionizing radiation49. ATM is an established pancreatic cancer susceptibility gene and recent evidence suggests that ATM germline mutations are among the most common found in familial and sporadic PDAC patients8, 9, 11, 50. PTCH1 and SUFU are both components of the Hedgehog signaling pathway. PTCH1 is a transmembrane protein that suppresses Hedgehog signaling when not bound to ligand, while SUFU is a cytoplasmic protein that inhibits Hedgehog signaling through binding of GLI transcription factors51. Germline mutations in PTCH1 and SUFU are implicated in Gorlin syndrome and predisposition to childhood medulloblastoma5254. PTCH1 and SUFU are intriguing candidate pancreatic cancer susceptibility genes as aberrant Hedgehog signaling has been implicated in pancreatic tumor development. Specifically, over-expression of SHH is observed in over 70% of pancreatic tumors and results in autocrine mediated changes to the tumor-microenvironment55, 56. Furthermore, PTCH1 and SUFU can be somatically mutated in PDAC11, 5759. Additional large cohort studies of IPMN and PDAC patients will be needed to determine the prevalence of PTCH1 and SUFU germline mutations and risk of tumor development.

Interestingly, surgically resected IPMN patients with a germline mutation in a pancreatic cancer susceptibility gene were significantly more likely to have concurrent invasive pancreatic carcinoma than patients without a germline mutation (Table 4). The majority of patients with a germline mutation in a pancreatic cancer susceptibility gene and invasive carcinoma did not have a reported family history of pancreatic cancer (4 of 5 patients) or personal cancer history (5 of 5 patients). This may indicate that the presence of a germline mutation in a pancreatic cancer susceptibility gene is an independent risk factor for progression to PDAC. Prospective studies, however, are necessary to determine the magnitude of any increased risk60.

Recent studies have suggested that knowledge of germline status in PDAC patients may be of limited personal utility, except for guiding use of PARP-1 inhibitors and immunotherapies in patients with defects in homology-directed and mismatch DNA repair respectively1517. Knowledge of germline status in patients with an IPMN, however, may be advantageous. Specifically, IPMN patients with a germline mutation may warrant additional surveillance to diagnose pancreatic and extra-pancreatic tumors, as is the case for germline mutation carriers with a family history of PDAC61, 62. Additional prospective studies are needed to confirm that additional screening in this patient population improves early diagnosis rates and patient outcomes.

Our study has several limitations. First, this is a retrospective study of patients with surgically resected IPMN. While this assured that all IPMNs were histologically confirmed, these patients are a subset of all patients with IPMN. Specifically, our study included patients with IPMNs advanced enough to warrant surgery and therefore, may be more likely to have already or in the future develop PDAC. Assessment of unselected patients is necessary to determine the clinical utility of stratification by germline mutation status in patients with IPMN that have not yet undergone surgical resection. Second, while we present the largest characterization of hereditary cancer predisposition genes in IPMN patients to date, our sample size is too small to detect associations with germline mutations that are a rare cause of IPMN or PDAC. Third, we used publicly available data from ExAC for controls as a large dataset of similarly sequenced controls was not available. Variant data from ExAC samples was similarly annotated and analyzed to IPMN cases, however, sequencing methodology was different, and this may result in batch effects that hinder analysis of gene associations. Fourth, only limited clinicopathologic data were available, therefore, associations between cancer-risk factors other than those presented in the study and germline mutation status could not be explored.

In conclusion, we characterized germline mutations in hereditary cancer predisposition genes in surgically resected IPMN patients. We found that germline mutations were most frequently identified in ATM and BRCA2 and that germline line mutations in ATM, PTCH1, and SUFU were significantly more common in patients with an IPMN than in ExAC controls. Furthermore, IPMN patients with a germline mutation in a pancreatic cancer susceptibility gene were significantly more likely to have concurrent invasive pancreatic carcinoma. Our study indicates that germline testing of IPMN patients is warranted and may have important implications for patient care.

Supplementary Material

1

Acknowledgments

Funding sources

This work was supported by: The Sol Goldman Pancreatic Cancer Research Center; Susan Wojcicki and Dennis Troper; The Lustgarten Foundation; The Rolfe Pancreatic Cancer Foundation; The Joseph C Monastra Foundation; The Gerald O Mann Charitable Foundation (Harriet and Allan Wulfstat, Trustees); NIH/NCI R00 CA190889 and NIH/NCI P50 CA62924.

Abbreviations

ATM

Ataxia telangiectasia mutated

bp

Base pair

BRCA1

breast cancer 1

BRCA2

breast cancer 2

BUB1B

BUB1 mitotic checkpoint serine/threonine kinase B

CDKN2A

cyclin-dependent kinase inhibitor 2A

CPA1

carboxypeptidase A1

ExAC

Exome Aggregation Consortium

FANCI

FA complementation group I

FANCM

FA complementation group M

FFPE

formalin fixed, paraffin-embedded

GLI1

GLI family zinc finger 1

IPMNs

intraductal papillary mucinous neoplasms

MAF

minor allele frequency

MLH1

mutL homolog 1

MSH2

mutS homolog 2

PDAC

pancreatic adenocarcinoma

PALB2

partner and localizer of BRCA2

PMS2

PMS1 homolog 2, mismatch repair system component

PARP-1

poly [ADP-ribose] polymerase-1

PRSS1

serine protease 1

PTCH1

patched 1

STK11

serine/threonine kinase 11

SUFU

SUFU negative regulator of hedgehog signaling

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflicts of interest

The authors declare no conflicts of interest.

References

  • 1.Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin 2017;67:7–30. [DOI] [PubMed] [Google Scholar]
  • 2.Amundadottir LT. Pancreatic Cancer Genetics. Int J Biol Sci 2016;12:314–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Canto MI, Almario JA, Schulick RD, et al. Risk of Neoplastic Progression in Individuals at High Risk for Pancreatic Cancer Undergoing Long-term Surveillance. Gastroenterology 2018;155:740–751e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Chen F, Roberts NJ, Klein AP. Inherited pancreatic cancer. Chin Clin Oncol 2017;6:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hu C, Hart SN, Bamlet WR, et al. Prevalence of Pathogenic Mutations in Cancer Predisposition Genes among Pancreatic Cancer Patients. Cancer Epidemiol Biomarkers Prev 2016;25:207–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Grant RC, Selander I, Connor AA, et al. Prevalence of germline mutations in cancer predisposition genes in patients with pancreatic cancer. Gastroenterology 2015;148:556–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Holter S, Borgida A, Dodd A, et al. Germline BRCA Mutations in a Large Clinic-Based Cohort of Patients With Pancreatic Adenocarcinoma. J Clin Oncol 2015;33:3124–9. [DOI] [PubMed] [Google Scholar]
  • 8.Shindo K, Yu J, Suenaga M, et al. Deleterious Germline Mutations in Patients With Apparently Sporadic Pancreatic Adenocarcinoma. J Clin Oncol 2017;35:3382–3390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hu C, Hart SN, Polley EC, et al. Association Between Inherited Germline Mutations in Cancer Predisposition Genes and Risk of Pancreatic Cancer. JAMA 2018;319:2401–2409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hu C, LaDuca H, Shimelis H, et al. Multigene Hereditary Cancer Panels Reveal High-Risk Pancreatic Cancer Susceptibility Genes. JCO Precision Oncology 2018:1–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Roberts NJ, Norris AL, Petersen GM, et al. Whole Genome Sequencing Defines the Genetic Heterogeneity of Familial Pancreatic Cancer. Cancer Discov 2016;6:166–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Tamura K, Yu J, Hata T, et al. Mutations in the pancreatic secretory enzymes CPA1 and CPB1 are associated with pancreatic cancer. Proc Natl Acad Sci USA 2018;115:4767–4772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Tanaka M Intraductal Papillary Mucinous Neoplasm of the Pancreas as the Main Focus for Early Detection of Pancreatic Adenocarcinoma. Pancreas 2018;47:544–550. [DOI] [PubMed] [Google Scholar]
  • 14.Hruban RH, Canto MI, Goggins M, et al. Update on familial pancreatic cancer. Adv Surg 2010;44:293–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Villarroel MC, Rajeshkumar NV, Garrido-Laguna I, et al. Personalizing cancer treatment in the age of global genomic analyses: PALB2 gene mutations and the response to DNA damaging agents in pancreatic cancer. Mol Cancer Ther 2011;10:3–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.O’Reilly EM, Lee JW, Lowery MA, et al. Phase 1 trial evaluating cisplatin, gemcitabine, and veliparib in 2 patient cohorts: Germline BRCA mutation carriers and wild-type BRCA pancreatic ductal adenocarcinoma. Cancer 2018;124:1374–1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hruban RH, Goggins M, Parsons J, et al. Progression model for pancreatic cancer. Clin Cancer Res 2000;6:2969–72. [PubMed] [Google Scholar]
  • 19.Notta F, Chan-Seng-Yue M, Lemire M, et al. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature 2016;538:378–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Vogelstein B, Papadopoulos N, Velculescu VE, et al. Cancer genome landscapes. Science 2013;339:1546–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Makohon-Moore AP, Matsukuma K, Zhang M, et al. Precancerous neoplastic cells can move through the pancreatic ductal system. Nature 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Pittman ME, Rao R, Hruban RH. Classification, Morphology, Molecular Pathogenesis, and Outcome of Premalignant Lesions of the Pancreas. Arch Pathol Lab Med 2017;141:1606–1614. [DOI] [PubMed] [Google Scholar]
  • 23.Kleeff J, Korc M, Apte M, et al. Pancreatic cancer. Nat Rev Dis Primers 2016;2:16022. [DOI] [PubMed] [Google Scholar]
  • 24.Laffan TA, Horton KM, Klein AP, et al. Prevalence of unsuspected pancreatic cysts on MDCT. AJR Am J Roentgenol 2008;191:802–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lennon AM, Wolfgang CL, Canto MI, et al. The early detection of pancreatic cancer: what will it take to diagnose and treat curable pancreatic neoplasia? Cancer Res 2014;74:3381–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Tanaka M, Chari S, Adsay V, et al. International consensus guidelines for management of intraductal papillary mucinous neoplasms and mucinous cystic neoplasms of the pancreas. Pancreatology 2006;6:17–32. [DOI] [PubMed] [Google Scholar]
  • 27.Vege SS, Ziring B, Jain R, et al. American gastroenterological association institute guideline on the diagnosis and management of asymptomatic neoplastic pancreatic cysts. Gastroenterology 2015;148:819–22; [DOI] [PubMed] [Google Scholar]
  • 28.Tanaka M, Fernandez-del Castillo C, Adsay V, et al. International consensus guidelines 2012 for the management of IPMN and MCN of the pancreas. Pancreatology 2012;12:183–97. [DOI] [PubMed] [Google Scholar]
  • 29.Tanaka M, Fernandez-Del Castillo C, Kamisawa T, et al. Revisions of international consensus Fukuoka guidelines for the management of IPMN of the pancreas. Pancreatology 2017;17:738–753. [DOI] [PubMed] [Google Scholar]
  • 30.Matthaei H, Norris AL, Tsiatis AC, et al. Clinicopathological characteristics and molecular analyses of multifocal intraductal papillary mucinous neoplasms of the pancreas. Ann Surg 2012;255:326–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kang MJ, Jang JY, Lee KB, et al. Long-term prospective cohort study of patients undergoing pancreatectomy for intraductal papillary mucinous neoplasm of the pancreas: implications for postoperative surveillance. Ann Surg 2014;260:356–63. [DOI] [PubMed] [Google Scholar]
  • 32.Pea A, Yu J, Rezaee N, et al. Targeted DNA Sequencing Reveals Patterns of Local Progression in the Pancreatic Remnant Following Resection of Intraductal Papillary Mucinous Neoplasm (IPMN) of the Pancreas. Ann Surg 2017;266:133–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Martinez de Juan F, Reolid Escribano M, Martinez Lapiedra C, et al. Pancreatic adenosquamous carcinoma and intraductal papillary mucinous neoplasm in a CDKN2A germline mutation carrier. World J Gastrointest Oncol 2017;9:390–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sato N, Rosty C, Jansen M, et al. STK11/LKB1 Peutz-Jeghers gene inactivation in intraductal papillary-mucinous neoplasms of the pancreas. Am J Pathol 2001;159:2017–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lubezky N, Ben-Haim M, Lahat G, et al. Intraductal papillary mucinous neoplasm of the pancreas: associated cancers, family history, genetic predisposition? Surgery 2012;151:70–5. [DOI] [PubMed] [Google Scholar]
  • 36.Canto MI, Goggins M, Hruban RH, et al. Screening for early pancreatic neoplasia in high-risk individuals: a prospective controlled study. Clin Gastroenterol Hepatol 2006;4:766–81; [DOI] [PubMed] [Google Scholar]
  • 37.Shi C, Klein AP, Goggins M, et al. Increased Prevalence of Precursor Lesions in Familial Pancreatic Cancer Patients. Clin Cancer Res 2009;15:7737–7743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Panic N, Capurso G, Attili F, et al. Risk for Colorectal Adenomas Among Patients with Pancreatic Intraductal Papillary Mucinous Neoplasms: a Prospective Case-Control Study. J Gastrointestin Liver Dis 2015;24:445–50. [DOI] [PubMed] [Google Scholar]
  • 39.Khan S, Sclabas G, Reid-Lombardo KM. Population-based epidemiology, risk factors and screening of intraductal papillary mucinous neoplasm patients. World J Gastrointest Surg 2010;2:314–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Reid-Lombardo KM, Mathis KL, Wood CM, et al. Frequency of extrapancreatic neoplasms in intraductal papillary mucinous neoplasm of the pancreas: implications for management. Ann Surg 2010;251:64–9. [DOI] [PubMed] [Google Scholar]
  • 41.Benarroch-Gampel J, Riall TS. Extrapancreatic malignancies and intraductal papillary mucinous neoplasms of the pancreas. World J Gastrointest Surg 2010;2:363–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 2009;25:1754–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.McKenna A, Hanna M, Banks E, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 2010;20:1297–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res 2010;38:e164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Genomes Project C, Auton A, Brooks LD, et al. A global reference for human genetic variation. Nature 2015;526:68–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lek M, Karczewski KJ, Minikel EV, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 2016;536:285–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Robinson JT, Thorvaldsdottir H, Winckler W, et al. Integrative genomics viewer. Nat Biotechnol 2011;29:24–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.LaRusch J, Whitcomb DC. Genetics of pancreatitis. Curr Opin Gastroenterol 2011;27:467–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lavin MF. Ataxia-telangiectasia: from a rare disorder to a paradigm for cell signalling and cancer. Nat Rev Mol Cell Biol 2008;9:759–69. [DOI] [PubMed] [Google Scholar]
  • 50.Roberts NJ, Jiao Y, Yu J, et al. ATM mutations in patients with hereditary pancreatic cancer. Cancer Discov 2012;2:41–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wu F, Zhang Y, Sun B, et al. Hedgehog Signaling: From Basic Biology to Cancer Therapy. Cell Chem Biol 2017;24:252–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wolter M, Reifenberger J, Sommer C, et al. Mutations in the human homologue of the Drosophila segment polarity gene patched (PTCH) in sporadic basal cell carcinomas of the skin and primitive neuroectodermal tumors of the central nervous system. Cancer Res 1997;57:2581–5. [PubMed] [Google Scholar]
  • 53.Pastorino L, Ghiorzo P, Nasti S, et al. Identification of a SUFU germline mutation in a family with Gorlin syndrome. Am J Med Genet A 2009;149A:1539–43. [DOI] [PubMed] [Google Scholar]
  • 54.Waszak SM, Northcott PA, Buchhalter I, et al. Spectrum and prevalence of genetic predisposition in medulloblastoma: a retrospective genetic study and prospective validation in a clinical trial cohort. Lancet Oncol 2018;19:785–798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Thayer SP, di Magliano MP, Heiser PW, et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 2003;425:851–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Rhim AD, Oberstein PE, Thomas DH, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 2014;25:735–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Biankin AV, Waddell N, Kassahn KS, et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012;491:399–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Waddell N, Pajic M, Patch AM, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015;518:495–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Forbes SA, Beare D, Boutselakis H, et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res 2017;45:D777–D783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mukewar SS, Sharma A, Phillip N, et al. Risk of Pancreatic Cancer in Patients With Pancreatic Cysts and Family History of Pancreatic Cancer. Clin Gastroenterol Hepatol 2018;16:1123–1130e1. [DOI] [PubMed] [Google Scholar]
  • 61.Canto MI, Hruban RH, Fishman EK, et al. Frequent detection of pancreatic lesions in asymptomatic high-risk individuals. Gastroenterology 2012;142:796–804; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Vasen H, Ibrahim I, Ponce CG, et al. Benefit of Surveillance for Pancreatic Cancer in High-Risk Individuals: Outcome of Long-Term Prospective Follow-Up Studies From Three European Expert Centers. J Clin Oncol 2016;34:2010–9. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

1

RESOURCES