Next Article in Journal
Insights into Epigenetic Changes Related to Genetic Variants and Cells-of-Origin of Pancreatic Neuroendocrine Tumors: An Algorithm for Practical Workup
Next Article in Special Issue
Inhibition of KRAS, MEK and PI3K Demonstrate Synergistic Anti-Tumor Effects in Pancreatic Ductal Adenocarcinoma Cell Lines
Previous Article in Journal
Molecular Divergence upon EGFR-TKI Resistance Could Be Dependent on the Exon Location of the Original EGFR-Sensitizing Mutation
Previous Article in Special Issue
A Genome-First Approach to Estimate Prevalence of Germline Pathogenic Variants and Risk of Pancreatic Cancer in Select Cancer Susceptibility Genes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Clinical Significance of Germline Pathogenic Variants among 51 Cancer Predisposition Genes in an Unselected Cohort of Italian Pancreatic Cancer Patients

by
Alberto Puccini
1,2,
Marta Ponzano
3,
Bruna Dalmasso
4,
Irene Vanni
2,4,
Annalice Gandini
1,2,
Silvia Puglisi
1,2,
Roberto Borea
1,2,
Malvina Cremante
1,2,
William Bruno
2,4,
Virginia Andreotti
4,
Eleonora Allavena
2,
Valentino Martelli
1,2,
Fabio Catalano
1,2,
Massimiliano Grassi
1,2,
Maria Laura Iaia
1,2,
Chiara Pirrone
1,2,
Alessandro Pastorino
1,
Giuseppe Fornarini
1,
Stefania Sciallero
1,†,
Paola Ghiorzo
2,4,*,† and
Lorenza Pastorino
2,4,†
add Show full author list remove Hide full author list
1
Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
2
Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, V.le Benedetto XV 6, 16132 Genoa, Italy
3
Department of Health Sciences (DISSAL), University of Genova, 16132 Genova, Italy
4
Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, L.go Rosanna Benzi X, 16132 Genoa, Italy
*
Author to whom correspondence should be addressed.
These authors jointly coordinated this study.
Submission received: 8 August 2022 / Revised: 29 August 2022 / Accepted: 2 September 2022 / Published: 13 September 2022
(This article belongs to the Special Issue Molecular Genetics of Pancreatic Cancer and Translational Challenges)

Abstract

:

Simple Summary

Multigene germline panel testing data, extended beyond BRCA, in unselected pancreatic cancer patients, are missing in the Italian population. We aimed here to determine the prevalence and impact of pathogenic variants in 51 pancreatic cancer candidate susceptibility genes in an unselected cohort of Italian pancreatic cancer patients. We found that 17% were carriers. CDKN2A was the most frequently mutated gene, followed by BRCA2 and ATM. Carriers showed better overall survival. A total of 41% of them had no family history. All CDKN2A carriers were older than 50 years, and BRCA1/2 carriers were younger than 70 years. CDKN2A and ATM should be added to current BRCA1/2 testing independently of family history in our population.

Abstract

Multigene germline panel testing is recommended for Pancreatic Cancer (PC) patients; however, for non-BRCA1/2 genes, the clinical utility is unclear. A comprehensive multi-gene assessment in unselected Italian PC patients is missing. We evaluated the prevalence and impact of Pathogenic Variants (PV) in 51 PC susceptibility genes in a real-world series of 422 Italian PC patients unselected for Family History (FH), compared the clinical characteristics and conducted survival analyses. 17% of patients had PVs (70/422), mainly in BRCA1/2 (4.5%, all <70 y), CDKN2A (4.5%, all >50 y), ATM (2.1%). PV carriers were younger (64 vs. 67; p = 0.02) and had more frequent personal/FH of PC, melanoma and breast/ovarian cancer (all p < 0.05). The Overall Survival (OS) was longer in patients carrying PVs (HR 0.78; p = 0.090), comprising ATM carriers (HR 0.33; p = 0.054). In the oxaliplatin-treated subset, PV carriers showed better control of the disease, although this was not statistically significant (67% vs. 56%). CDKN2A, BRCA2 and ATM were the most frequently altered genes. ATM PVs were positively associated with OS in 41% of PV carriers, 60% of whom carried CDKN2A, BRCA2 or ATM PVs, had negative FH and would have been missed by traditional referral. Thus, CDKN2A and ATM should be added to BRCA1/2 testing regardless of FH.

Graphical Abstract

1. Introduction

Exocrine Pancreatic Cancer (PC) is one of the deadliest solid tumors, with an average 5-year survival rate of 5–10% [1]. In 2020, 495,773 new cases and 466,003 deaths were observed globally (4.5% of all deaths caused by cancer) [2,3,4], and the incidence is expected to increase [5]. Pancreatic cancer remains one of the few cancers in which both early diagnosis and effective therapeutic processes have been lacking in recent years. Surgical resection is the only potentially curative treatment; however, less than 20% of patients present with resectable disease at the time of diagnosis [6,7].
Up to 10% of pancreatic cancer patients have familial inheritance [8]: about 3% develop pancreatic cancer in the context of hereditary cancer syndromes, and another 7% are classified as Familial Pancreatic Cancer (FPC), a definition attributed to individuals who have two or more first-degree relatives with pancreatic cancer.
Hereditary pancreatic cancer syndromes include Hereditary Breast and Ovarian Cancer syndrome (HBOC), Peutz–Jeghers syndrome (SPJ), Familial Atypical Multiple Mole Melanoma (FAMMM)/melanoma-pancreatic cancer syndrome, Lynch syndrome (or Hereditary Non Polyposis Colorectal Carcinoma [HNPCC]), Familial Adenomatous Polyposis (FAP), ataxia-telangiectasia (ATM) and Hereditary Pancreatitis (HP). These hereditary cancer syndromes account for 10–15% of hereditary pancreatic cancers, while a genetic explanation for most FPCs has yet to be identified [9,10,11].
However, there are no established screening procedures for high-risk unaffected cases or for the general population, and it is unclear whether surveillance programs would provide clinical benefits [12]. Despite this lack of an internationally validated surveillance program for high-risk individuals, international guidelines (e.g., the National Comprehensive Cancer Network (NCCN) guidelines) recommend multigene germline testing for any patient with a confirmed PC diagnosis [13].
The implementation of this recommendation is hampered by the uncertainty around the clinical utility of multigene testing in pancreatic cancer patients, particularly for genes other than BRCA1/2. Indeed, Multi Gene Panel (MGP) testing results in a high rate of Pathogenic Variants (PVs), whose spectrum of cancer risks and penetrance is often not clearly defined. In addition, MGP testing generates high rates of Variants of Unknown Significance (VUS). These issues concern the medical community, as ambiguous findings may lead to unnecessary patient anxiety and unwarranted interventions [14].
In addition, the widespread availability of Next-Generation Sequencing (NGS) technologies, which has resulted in increased detection rate of PVs and VUS through MGP testing, has led to a paradigm shift also in cancer susceptibility testing [15]. Indeed, rather than submitting patients to pre-and post-genetic testing sessions, we were called to perform mainstream, oncologist-led, family history-independent genetic testing and refer for genetic counseling only those carrying a germline variant or with a strong clinical suspect of hereditary cancer syndrome [16].
For these reasons, while previously no more than 10% of PC cases were considered due to heritable factors, only a subset of which was explained by germline PVs in high-risk genes, recent sequencing efforts have revealed that as many as 15–20% of PC patients unselected for Family History (FH) bear PVs [17,18]. The use of MGP testing has also been implemented to detect druggable PVs (NCCN Guidelines 2022). The results from the POLO trial [19,20] confirmed the proof of principle of anti-PARP (Poly ADP-ribose polymerase) efficacy in PC patients harboring germline PVs in the BRCA1/2 genes.
In addition to ovarian [21], breast [22] and prostate cancer patients [23], the efficacy shown in this study extends the population to be tested and counseled for BRCA PVs to include PC patients. In addition, some other above mentioned hereditary syndromes associated with PC (i.e., Lynch Syndrome) are known to be caused by actionable mutations [24]. However, oncologists tend to focus on cancer treatment and overlook the implications of genetic counseling and diagnosis of hereditary syndromes.
Thus, the POLO trial’s results have two main implications: (a) the need to select BRCA germline positive patients for personalized therapy and (b) the need to adequately counsel them for hereditary cancer syndromes. Of note, the US Food and Drug Administration (FDA), as well as the European Medicines Agency (EMA), approved Olaparib in this setting of patients based on POLO trial in 2019 [25] and 2020 [26] respectively, while the Italian Medicines Agency (AIFA) recently withdrew Olaparib from the approved treatments for PCs.
The POLO study also confirmed the well-known platinum sensitivity of patients carrying germline PVs causing DNA Damage Repair—Homologous Recombination Deficiency (DDR-HRD), such as BRCA1/2 [27]. However, a comprehensive analysis by MGP in unselected Italian PC patients is lacking. Preliminary data from different cohorts of unselected Italian PC patients focusing on CDKN2A and BRCA1/2 showed a high prevalence of CDKN2A PVs, regardless of familial status [28] and of BRCA1/2 PVs—these latter being observed only in patients < 74 y [29].
This study aimed to evaluate the prevalence and impact on outcomes of PVs in 51 PC susceptibility genes in a retrospective/prospective series of 422 Italian PC patients recruited at our Institution and unselected for FH.

2. Materials and Methods

2.1. Patients Selection and Characteristic

From 2020 to 2022, we recruited a consecutive series of 111 PC patients referred to our center for molecular analysis. Moreover, we retrospectively included an additional series of 311 non-consecutive PC patients who underwent molecular analysis at our center. Patients with neuroendocrine pancreatic tumors were excluded. All patients (N = 422) were unselected for age, FH or personal history, and there were no differences in terms of patients’ characteristics (age, family, personal history and prevalence of PVs) between retrospective and prospective groups, except for the stage and Overall Survival (OS).
This may be explained by stage migration and better management of patients in recent years. All patients signed an informed consent according to local ethics committee protocols and answered a standardized family history questionnaire on the presence and type of cancers in the family. One hundred twenty-two patients have been described in previous publications; however, the analysis was limited to CDKN2A or BRCA1-2 [28,30], as well as Mismatch Repair (MMR) genes for a subset of high-risk patients [30,31]. In addition, 31 patients, either positive for CDKN2A PVs or negative but considered high-risk because of PC family history, had been previously tested by Whole-Exome Sequencing (WES) [32] and were re-tested here by MGP.

2.2. Gene Panel

Next-Generation Sequencing (NGS) was performed for all 422 patients using an Ion Custom Panel (125–275 bp amplicon target sizes; 355 kb; 1266 amplicons), including 51 genes involved in DDR, PC susceptibility and hereditary pancreatitis (APC, ARID1A, ARID1B, ARID2, ATM, ATR, ATRX, BAP1, BARD1, BMPR1A, BRCA1, BRCA2, BRIP1, CDC73, CDK12, CDK4, CDKN2A, CHEK1, CHEK2, COL7A1, CPA1, EPCAM, ERCC4, FAM175A, FANCA, FANCB, FANCC, FANCD2, FANCE, FANCF, FANCG, FANCL, MLH1, MRE11, MSH2, MSH6, NBN, PALB2, PMS2, PRSS1, RABL3, RAD50, RAD51, RAD51B, RAD51C, RAD51D, RET, SPINK1, STK11, SUFU and TP53) according to the recent literature [33,34].
The NGS libraries were constructed using the Ion AmpliSeq Kit for Chef DL8 (Thermo Fisher Scientific, Waltham, MA, USA), according to the manufacturer’s protocol starting from 20 ng of genomic DNA. According to the manufacturer, the libraries were sequenced with an Ion Torrent S5 GeneStudio Plus system using 530 Chip (Thermo Fisher Scientific). The S5 sequencing data were analyzed by the Ion Torrent Software Suite (Thermo Fisher Scientific) using the plugin Variant Caller and Ion Reporter (Thermo Fisher Scientific). The filtered variants were analyzed and classified according to the American College of Medical Genetics and Genomics (ACMG) classification [35]. Only ACMG class 4 (Likely Pathogenic) and 5 (Pathogenic) variants were considered as PVs in this study.

2.3. Statistical Analysis

Descriptive characteristics were reported as mean (standard deviation) or absolute frequency (relative frequency). Patients carrying PVs were compared to the non-carriers using T-test for continuous variables and Chi-squared or Fisher’s exact test for categorical variables. Univariable Cox proportional hazards models were used to identify characteristics associated with prognosis in terms of overall survival. Subsequently, we presented multivariable Cox proportional hazards models adjusting for confounders. Additionally, as a sensitivity analysis, age-adjusted Cox proportional hazards models were performed to specifically assess the impact of each type of mutation.
Within the subsample of patients who received oxaliplatin-based treatment, the association between PVs and disease progression was evaluated by a Chi-squared test. Finally, an age-adjusted Cox proportional hazards model was performed to compare responding and not-responding patients. All statistical analyses were conducted using Stata version 16.0 (Stata Corporation, College Station, TX, USA), and p-values < 0.05 were considered statistically significant.

3. Results

We recruited 422 patients belonging to a real-world series of patients diagnosed with Pancreatic cancer, unselected for family history and analyzed them using a multigene panel containing 51 genes. Table 1 shows the patients’ characteristics according to the germline status.
Overall, 52% were females, and the mean age was 67 years (range 30–92). As expected, germline PV carriers were significantly younger than non-carriers (p-value = 0.0254). Overall, 13% of patients had a first or second-degree relative with PC. A family history of PC was more frequent in carriers than in non-carriers (26% vs. 11%; p = 0.001); the same pattern was confirmed in cases with a personal and/or family history of either breast and ovarian cancer (36% vs. 14%, p < 0.001), melanoma (19% vs. 5%; p < 0.01) and personal history of other tumors (32% vs. 13%, p < 0.001).

3.1. Prevalence of Pathogenic Variants in Pancreatic Cancer Susceptibility Genes: Correlation with Family History and Age at Diagnosis

Seventy out of 422 enrolled patients (17%) carried one or more pathogenic variant (PV) in at least one of the 51 genes included in the panel (Table 1). CDKN2A was the most frequently mutated gene (19/422; 4.5%), confirming the founder p.Gly101Trp as the most frequent PV in our series (11/19 = 57.9%) [36]. For this reason, CDKN2A in our cohort shows a higher frequency compared to the literature data [37] (Figure 1).
BRCA2 (13/422 = 3.1%) was the second most frequently mutated gene. BRCA1/2 positive patients (19/422; 4.5%) were as frequent as those carrying CDKN2A PV/LPVs. The third most frequently mutated gene was ATM since PV/LPVs were detected in nine patients (2.1%). Altogether, PV/LPVs in these four genes were 50% of all mutations in our cohort (37/74). Notably, 41% (29/70) of patients carrying PV/LPVs had no pancreatic, breast or melanoma personal and/or family history (Table 2).
The prevalence of PVs in HRD pathway-related genes (ATM, BRCA1/2, CHEK2, ERCC4, FANCA, FANCC, FANCG, NBN, RAD50 and RAD51) was 10.4%. The inclusion of HR-DDR pathway-related genes compared to BRCA1/2 alone more than doubled the prevalence of pathogenic variants (10.4% vs. 4.5%). Among these PVs, 41% (18/44) were found in sporadic patients.
Three patients showed multiple PVs: two sporadic PC patients had two (in BRCA2 and CHEK2) and three (in ATM, CDKN2A and NBN) pathogenic variants, respectively. The third, who developed both melanoma and PC, showed 2 PVs (in CDKN2A and NBN). Interestingly, two out of three patients carrying NBN PVs had a concomitant PV in a high penetrance predisposition gene (CDKN2A or ATM).
Seven PVs were found in CHEK2 (1.7%), four of whom had no FH. Five PVs (1.2%) were found in the COL7A1 gene; among these patients, only two showed a family history of PC and breast cancer. Finally, we detected two PVs in MSH2, in 2 patients with suspected Lynch Syndrome. No variants were identified in the PALB2 gene. The complete list of LPs and PVs is described in Supplementary Table S1.
As expected, PVs in BRCA1/2 were more frequent in subjects with a personal and/or family history for breast/ovarian cancer (12/76, 15.8%). However, a 1.8% frequency was observed even in sporadic subjects (5/283). CDKN2A PVs were frequent in patients with a family history of PC (8/55, 14.5%) and with a personal or family history of melanoma (6/30, 20%); however, the rate remains high (8/283, 2.8%) in sporadic subjects. Notably, ATM PVs frequency was 5.4% in patients with a family history of PC (3/55) and 3.3% in patients with personal or family history of melanoma (1/30). ATM PVs mutation frequency was as high as that of BRCA-2 (1.8%) in sporadic patients.
Overall, 11.3% of PV were found in sporadic patients (32/283) and would have been missed if we tested patients based only on family history (Table 2). When we analyzed age at diagnosis comparing patients according to specific PVs, we found that the overall frequency of PVs was 22.2% among patients ≤50 y, 24.4% among patients 51–60 y, 17.5% among patients 61–70 y and 11.6% for those who were >70 y. The highest frequency of BRCA1/2 PV was found among the youngest patients (≤50, 13.9%), while no PVs were found in patients older than 70.
Conversely, the highest CDKN2A mutation rate was found in patients aged 61–70 y (6.3%) and was 3.8% in patients > 70 y. No PVs in CDKN2A were found in patients younger than 50 y (Supplementary Table S2). Overall, patients with BRCA1/2 genes were younger (mean age = 58) than CDKN2A patients (mean age = 67) (Supplementary Table S2).

3.2. Impact of Germline Pathogenic Variants on Patients’ Survival

Table 3 summarizes patients characteristics according to OS. Median follow-up was 9.7 months (interquartile range: 4.3–21,25) and median OS was 11.5 months (range 10.4–13.3). In univariable analysis, as expected, 10-years increase in age was associated with worse prognosis (HR 1.18; 95% CI 1.06–1.32; p = 0.004), as well as metastatic disease compared to resectable stage (HR 3.20; 95% CI 2.38–4.29; p < 0.001). These results were confirmed in multivariable analysis (age: HR 1.18; 95% CI 1.06–1.33; p = 0.004) (metastatic disease: HR 3.39; 95% CI 2.51–4.57; p < 0.001).
Patients carrying any PVs showed better OS compared to non-carriers in the univariable analysis (HR 0.78; 95% CI 0.59–1.04; p = 0.090) and was confirmed in the multivariable analysis (HR 0.81; 95% CI 0.61–1.09; p = 0.160). In the sensitivity analysis we also evaluated the impact of PVs in single gene and we found that all nine patients carrying ATM PVs had a better prognosis compared to non-carriers in terms of median OS (15 vs. 11 months; age-adjusted HR 0.33; 95% CI 0.10–1.02; p = 0.054). No significant differences were observed between patients with PVs in other genes and non-carriers.

3.3. Impact of Germline Pathogenic Variants on Response to Oxaliplatin

A total of 106 patients received oxaliplatin-based treatment: 25 (34%) in the neoadjuvant and 81 (76%) in the first- or second-line metastatic setting. Overall, in the neoadjuvant setting, 24% (N = 6) had a Progression of Disease (PD) as the Best Response (BR) to oxaliplatin, 28% (N = 7) had a partial response and 32% (N = 8) had stable disease. In the metastatic setting, 42% (N = 34) had a PD as the best response to oxaliplatin, 21% (N = 17) had a partial response, and 30% (N = 24) had a stable disease. Patients carrying a PV more frequently showed a good response to treatment (67%) compared to non-carriers (56%) but differences were not statistically significant (p = 0.381). When we focused on patients with BRCA or ATM PVs, all nine patients showed control of disease as BR to oxaliplatin.

4. Discussion

We analyzed a panel of 51 genes in a retrospective/prospective cohort of 422 Italian PC patients unselected for family history, stage or age of onset. To the best of our knowledge, this is the largest cohort of unselected PC Italian patients analyzed with a MGP comprehensive of all PC susceptibility genes described to date. Overall, 70 PVs (17%) were found, a relatively high frequency compared to similar studies. However, due to the heterogeneity in cohort selection and dissimilar sequencing approaches across studies, the prevalence of PVs in PC susceptibility genes cannot be easily quantified, ranging from 5% to nearly 20% in different studies and reviews [18,34,37,38,39,40,41,42].
Here, we confirm in an extended cohort of Italian PC patients, the high CDKN2A PV/LPV rate previously described [28,43], finding a total CDKN2A mutation rate of 4.5% (19/422), as high as the one conferred by BRCA1 and BRCA2 together (BRCA1 n = 6/422 and BRCA2 13/422). According to the literature, CDKN2A germline variants in patients unselected for FH range from 0.1 to 2.6% [37,44,45,46]. In our study, the rate increased to 14.5% (n = 8/55) and 20% (n = 6/30) in subjects with familial PC or with a personal and/or FH of melanoma, respectively.
The latest frequency confirms recent findings from an extended melanoma family study in the Italian population showing a 12.4% of PVs in melanoma families with PC cases [47]. However, the mutation rate remains high (2.8%) even in sporadic subjects. This may reflect the presence of founder CDKN2A PVs in our population [36].
As expected, BRCA1 and BRCA2 PV rate nearly doubled in cases with a FH of PC nearly compared to sporadic subjects (3.6% vs. 1.7%), while it ranged from 1.7% to 15.8% in subjects with a personal or family history of breast/ovarian cancer (Table 2). Again, these data confirm previously observed BRCA1/2 mutation rates in Italian families with history of both breast/ovarian and PC [40,41].
Carriers of BRCA1/2 PVs showed a lower age of onset compared to CDKN2A carriers. While CDKN2A PVs are distributed only in the age categories above 50 years, BRCA1/2 are the most common PVs in the age category below 50 years (13.9%). In our cohort, the oldest patient carrier of a germline BRCA1/2 PVs was 70 years old. This supports the results obtained in a different unselected cohort of Italian PC patients tested for BRCA1/2 only, showing that BRCA1/2 mutations were found in patients < 74 y [29].
ATM was the first most frequently mutated gene in the age category between 51 and 60 years and the third mutated gene overall (2.13%). Interestingly, ATM PV/LPVs frequency was second to CDKN2A only in patients with a family history of PC (5.4%) and was found at a frequency of 3.3% in patients with familiarity for melanoma. These data also confirm recent figures observed for ATM germline PVs in melanoma cases [47,48,49].
Overall, CHEK2 and COL7A1 PVs were found with a frequency above 1%. Further investigation will be needed to elucidate their role in pancreatic cancer predisposition. However, our findings are in line with those from the PAN Cancer [34]. A recent paper found that the combined frequency of heterozygous carriers of CHEK2 in the general population was 1.2% [50]. Interestingly, PVs in TP53 and MMR genes were below 1% and were found in patients with a personal and/or FH of breast/ovarian and colon cancer, respectively.
In our study, we observed that patients carrying any PVs had a better OS compared to non-carriers (HR = 0.66; 95% CI 0.47–0.93; p = 0.017). However, when we focused on PVs in specific genes, only patients carrying ATM PVs (N = 9) showed better OS compared to WT patients (N = 345) (HR = 0.33). Our results confirm previous reports that suggested ATM alterations as prognostic for improved outcomes in patients with PC [51].
Despite efforts in extensive molecular profiling in PC for actionable genetic alterations or pathways, targeted therapy showed to provide limited clinical benefit for patients with this disease to date [52]. One exception is alterations in DNA damage repair (DDR) genes, especially BRCA1/2, since BRCA-associated tumors, including breast, prostate and ovarian tumors, respond to poly–(adenosine diphosphate–ribose) polymerase (PARP) inhibitors and platinum-based therapy.
However, the efficacy of PARP inhibitors in DDR-related genes other than BRCA is less clear. Recent two phase II nonrandomized clinical trials showed that Olaparib was well tolerated. However, it showed limited antitumor activity in patients with advanced, platinum-sensitive PC with DDR-genetic alterations, most of them carried ATM alterations [53]. Unfortunately, no patients with ATM alterations were treated with PARP inhibitors in our cohort. However, we observed a trend in better disease control in PV carriers treated with oxaliplatin, although not statistically significant.
More data in larger cohorts are needed to confirm these results. Although the prevalence of DNA MMR is <1%, these patients may benefit from immunotherapy [24], thus we believe that evaluation of MMR system (somatic and/or germline) is crucial in PC patients as well as in all other solid tumor types [54]. BRCA1/2, CDKN2A and ATM, account for an overall PV/LPV prevalence of 11.1% in our cohort.
Had we selected PC patients for genetic testing according to their personal and family history suggestive for familial PC or hereditary cancer syndromes, we would have missed 41% of patient carriers of PVs in candidate gens. Sixty% of them were carriers of PVs in CDKN2A, BRCA2 and ATM. At least CDKN2A and ATM, should be added to BRCA1/2 testing by MGP independently of FH.
These PVs are clinically actionable since they result in hereditary cancer syndromes for which international guidelines support surveillance aimed to reduce mortality among relatives. Although such guidelines are not of proven efficacy for PC mortality reduction, identification of probands may still reduce mortality for other cancers through ‘ad hoc’ prevention protocols. However, several studies and meta-analyses have shown that the diagnostic yield of pre-malignant or malignant lesions (including earlier stage) in high risk individuals undergoing screening/surveillance is much higher than the lifetime risk of PC in the general population and this might be considered a surrogate for mortality reduction [55,56,57,58,59].
In Italy, the AISP (Italian Association for the Study of the Pancreas), developed a registry of families at high risk of PC to be included in surveillance protocols [59,60]. As previously mentioned, an extended MGP testing—beyond BRCA1 and 2—in all PC patients in not standard of care in Italy and an estimate of a real world MGP frequency in unselected PC patients was missing. The registry is currently recruiting families at high-risk because of family history or positive mutation testing. Our data support an extended MGP testing (at least to CDKN2A and ATM) to identify positive high-risk carriers to be included in registry surveillance protocols, also independently of family history.
We acknowledge that our study presents some limitations, mainly because of a non- consecutive and retrospective enrollment of part of the cohort. In addition, few data are available on treatments (oxaliplatin-based, anti-PARP and/or immunotherapy) in PV carriers. No correlation with somatic tumor samples was conducted. Moreover, stage migration and changes in patient management were observed during the long patient recruitment period.

5. Conclusions

In conclusion, our study showed that the universal use of MGP testing in PC patients allowed us to identify a high PV prevalence (17%). These data might address unmet clinical need to identify actionable mutations and provide the opportunity to identify well-known hereditary cancer syndromes. Finally, 40% of patients with PVs did not have suspected criteria within our cohort and would have been missed with standard selection criteria.
Thus, we propose extending the offer of genetic testing beyond BRCA1 and 2 at least to CDKN2A and ATM, independently of family history, with no age restrictions to identify mutations and actionable not only for target therapy in affected members but also for surveillance research protocols in carriers, including unaffected relatives.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/cancers14184447/s1, Table S1: List of Likely Pathogenic and Pathogenic Variants; Table S2: Overall mutation rate and related to a single gene.

Author Contributions

Conceptualization, methodology and writing—original draft preparation A.P. (Alberto Puccini), L.P., M.P., S.S. and P.G. All authors (B.D., I.V., A.G., S.P., R.B., M.C., W.B., V.A., E.A., V.M., F.C., M.G., M.L.I., C.P., A.P. (Alessandro Pastorino) and G.F.) made substantial contributions to this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from Lega Italiana per la Lotta contro i Tumori (LILT) 5 × 1000 IG 2019 to PG, Italian Ministry of Health (Ospedale Policlinico San Martino Ricerca Corrente and 5 × 1000 funds to PG), Associazione Ricerca Tumori Rari ed Ereditari (AR3) onlus to P.G. Alliance Against Cancer (REGISTRY OF THE EUROPEAN REFERENCE NETWORK ON RARE ADULT SOLID CANCER “EURACAN” [rif.21157; 21093; 21141]) to S.S.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board of IRCCS Policlinico San Martino (CER Liguria; approval #534/2020, approved on 23 November 2020).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

We wish to thank the participating patients and families, whose generosity and cooperation have made this study possible.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  3. Rawla, P.; Sunkara, T.; Gaduputi, V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J. Oncol. 2019, 10, 10–27. [Google Scholar] [CrossRef] [PubMed]
  4. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  5. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting Cancer Incidence and Deaths to 2030: The Unexpected Burden of Thyroid, Liver, and Pancreas Cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef]
  6. Ryan, D.P.; Hong, T.S.; Bardeesy, N. Pancreatic Adenocarcinoma. N. Engl. J. Med. 2014, 371, 1039–1049. [Google Scholar] [CrossRef]
  7. Janssen, Q.P.; O’Reilly, E.M.; van Eijck, C.H.J.; Groot Koerkamp, B. Neoadjuvant Treatment in Patients with Resectable and Borderline Resectable Pancreatic Cancer. Front. Oncol. 2020, 10, 41. [Google Scholar] [CrossRef]
  8. Welinsky, S.; Lucas, A.L. Familial Pancreatic Cancer and the Future of Directed Screening. Gut Liver 2017, 11, 761–770. [Google Scholar] [CrossRef]
  9. Syngal, S.; Brand, R.E.; Church, J.M.; Giardiello, F.M.; Hampel, H.L.; Burt, R.W. American College of Gastroenterology ACG Clinical Guideline: Genetic Testing and Management of Hereditary Gastrointestinal Cancer Syndromes. Am. J. Gastroenterol. 2015, 110, 223–262, quiz 263. [Google Scholar] [CrossRef]
  10. Hruban, R.H.; Canto, M.I.; Goggins, M.; Schulick, R.; Klein, A.P. Update on Familial Pancreatic Cancer. Adv. Surg. 2010, 44, 293–311. [Google Scholar] [CrossRef] [Green Version]
  11. Stoffel, E.M.; McKernin, S.E.; Khorana, A.A. Evaluating Susceptibility to Pancreatic Cancer: ASCO Clinical Practice Provisional Clinical Opinion Summary. J. Oncol. Pract. 2019, 15, 108–111. [Google Scholar] [CrossRef]
  12. Ohmoto, A.; Yachida, S.; Morizane, C. Genomic Features and Clinical Management of Patients with Hereditary Pancreatic Cancer Syndromes and Familial Pancreatic Cancer. Int. J. Mol. Sci. 2019, 20, 561. [Google Scholar] [CrossRef]
  13. Tempero, M.A.; Malafa, M.P.; Al-Hawary, M.; Behrman, S.W.; Benson, A.B.; Cardin, D.B.; Chiorean, E.G.; Chung, V.; Czito, B.; Del Chiaro, M.; et al. Pancreatic Adenocarcinoma, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2021, 19, 439–457. [Google Scholar] [CrossRef]
  14. Katz, S.J.; Ward, K.C.; Hamilton, A.S.; Abrahamse, P.; Hawley, S.T.; Kurian, A.W. Association of Germline Genetic Test Type and Results with Patient Cancer Worry After Diagnosis of Breast Cancer. JCO Precis. Oncol. 2018, 2, 1–8. [Google Scholar] [CrossRef]
  15. Hughes, K.S. Genetic Testing: What Problem Are We Trying to Solve? J. Clin. Oncol. 2017, 35, 3789–3791. [Google Scholar] [CrossRef]
  16. Colombo, N.; Huang, G.; Scambia, G.; Chalas, E.; Pignata, S.; Fiorica, J.; Van Le, L.; Ghamande, S.; González-Santiago, S.; Bover, I.; et al. Evaluation of a Streamlined Oncologist-Led BRCA Mutation Testing and Counseling Model for Patients with Ovarian Cancer. J. Clin. Oncol. 2018, 36, 1300–1307. [Google Scholar] [CrossRef]
  17. Athens, A.; Amacker-North, L.; Warsinske, K.; Kadakia, K.C.; Kim, E.S.; Salem, M.E.; Elrefai, S. Changing the Landscape of Germline Testing in Patients with Pancreatic Adenocarcinoma. JCO 2020, 38, 676. [Google Scholar] [CrossRef]
  18. Lowery, M.A.; Wong, W.; Jordan, E.J.; Lee, J.W.; Kemel, Y.; Vijai, J.; Mandelker, D.; Zehir, A.; Capanu, M.; Salo-Mullen, E.; et al. Prospective Evaluation of Germline Alterations in Patients with Exocrine Pancreatic Neoplasms. J. Natl. Cancer Inst. 2018, 110, 1067–1074. [Google Scholar] [CrossRef]
  19. Golan, T.; Hammel, P.; Reni, M.; Van Cutsem, E.; Macarulla, T.; Hall, M.J.; Park, J.-O.; Hochhauser, D.; Arnold, D.; Oh, D.-Y.; et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N. Engl. J. Med. 2019, 381, 317–327. [Google Scholar] [CrossRef]
  20. Golan, T.; Hammel, P.; Reni, M.; Van Cutsem, E.; Macarulla, T.; Hall, M.J.; Park, J.-O.; Hochhauser, D.; Arnold, D.; Oh, D.-Y.; et al. Overall Survival from the Phase 3 POLO Trial: Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. JCO 2021, 39, 378. [Google Scholar] [CrossRef]
  21. Banerjee, S.; Moore, K.N.; Colombo, N.; Scambia, G.; Kim, B.-G.; Oaknin, A.; Friedlander, M.; Lisyanskaya, A.; Floquet, A.; Leary, A.; et al. Maintenance Olaparib for Patients with Newly Diagnosed Advanced Ovarian Cancer and a BRCA Mutation (SOLO1/GOG 3004): 5-Year Follow-up of a Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. Lancet Oncol. 2021, 22, 1721–1731. [Google Scholar] [CrossRef]
  22. Tutt, A.N.J.; Garber, J.E.; Kaufman, B.; Viale, G.; Fumagalli, D.; Rastogi, P.; Gelber, R.D.; de Azambuja, E.; Fielding, A.; Balmaña, J.; et al. Adjuvant Olaparib for Patients with BRCA1- or BRCA2-Mutated Breast Cancer. N. Engl. J. Med. 2021, 384, 2394–2405. [Google Scholar] [CrossRef] [PubMed]
  23. De Bono, J.; Mateo, J.; Fizazi, K.; Saad, F.; Shore, N.; Sandhu, S.; Chi, K.N.; Sartor, O.; Agarwal, N.; Olmos, D.; et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020, 382, 2091–2102. [Google Scholar] [CrossRef] [PubMed]
  24. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch Repair Deficiency Predicts Response of Solid Tumors to PD-1 Blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef]
  25. FDA Approves Olaparib for GBRCAm Metastatic Pancreatic Adenocarcinoma. 27 December 2019. Available online: Https://www.Fda.Gov/Drugs/Resources-Information-Approved-Drugs/Fda-Approves-Olaparib-Gbrcam-Metastatic-Pancreatic-Adenocarcinoma#:~:Text=On%20December%2027%2C%202019%2C%20the,An%20FDA%2Dapproved%20test%2C%20whose (accessed on 1 July 2022).
  26. EMA Approval Olaparib in Pancreatic Cancer. 8 July 2020. Available online: Https://www.Ema.Europa.Eu/En/Medicines/Human/EPAR/Lynparza (accessed on 1 July 2022).
  27. Fogelman, D.; Sugar, E.A.; Oliver, G.; Shah, N.; Klein, A.; Alewine, C.; Wang, H.; Javle, M.; Shroff, R.; Wolff, R.A.; et al. Family History as a Marker of Platinum Sensitivity in Pancreatic Adenocarcinoma. Cancer Chemother. Pharmacol. 2015, 76, 489–498. [Google Scholar] [CrossRef]
  28. Ghiorzo, P.; Fornarini, G.; Sciallero, S.; Battistuzzi, L.; Belli, F.; Bernard, L.; Bonelli, L.; Borgonovo, G.; Bruno, W.; De Cian, F.; et al. CDKN2A Is the Main Susceptibility Gene in Italian Pancreatic Cancer Families. J. Med. Genet. 2012, 49, 164–170. [Google Scholar] [CrossRef]
  29. Peretti, U.; Cavaliere, A.; Niger, M.; Tortora, G.; Di Marco, M.C.; Rodriquenz, M.G.; Centonze, F.; Rapposelli, I.G.; Giordano, G.; De Vita, F.; et al. Germinal BRCA1-2 Pathogenic Variants (GBRCA1-2pv) and Pancreatic Cancer: Epidemiology of an Italian Patient Cohort. ESMO Open 2021, 6, 100032. [Google Scholar] [CrossRef]
  30. Ghiorzo, P.; Pensotti, V.; Fornarini, G.; Sciallero, S.; Battistuzzi, L.; Belli, F.; Bonelli, L.; Borgonovo, G.; Bruno, W.; Gozza, A.; et al. Contribution of Germline Mutations in the BRCA and PALB2 Genes to Pancreatic Cancer in Italy. Fam. Cancer 2012, 11, 41–47. [Google Scholar] [CrossRef]
  31. Gargiulo, S.; Torrini, M.; Ollila, S.; Nasti, S.; Pastorino, L.; Cusano, R.; Bonelli, L.; Battistuzzi, L.; Mastracci, L.; Bruno, W.; et al. Germline MLH1 and MSH2 Mutations in Italian Pancreatic Cancer Patients with Suspected Lynch Syndrome. Fam. Cancer 2009, 8, 547–553. [Google Scholar] [CrossRef]
  32. Yang, X.R.; Rotunno, M.; Xiao, Y.; Ingvar, C.; Helgadottir, H.; Pastorino, L.; van Doorn, R.; Bennett, H.; Graham, C.; Sampson, J.N.; et al. Multiple Rare Variants in High-Risk Pancreatic Cancer-Related Genes May Increase Risk for Pancreatic Cancer in a Subset of Patients with and without Germline CDKN2A Mutations. Hum. Genet. 2016, 135, 1241–1249. [Google Scholar] [CrossRef] [Green Version]
  33. Rainone, M.; Singh, I.; Salo-Mullen, E.E.; Stadler, Z.K.; O’Reilly, E.M. An Emerging Paradigm for Germline Testing in Pancreatic Ductal Adenocarcinoma and Immediate Implications for Clinical Practice: A Review. JAMA Oncol. 2020, 6, 764–771. [Google Scholar] [CrossRef]
  34. Huang, K.-L.; Mashl, R.J.; Wu, Y.; Ritter, D.I.; Wang, J.; Oh, C.; Paczkowska, M.; Reynolds, S.; Wyczalkowski, M.A.; Oak, N.; et al. Pathogenic Germline Variants in 10,389 Adult Cancers. Cell 2018, 173, 355–370.e14. [Google Scholar] [CrossRef]
  35. Richards, S.; Aziz, N.; Bale, S.; Bick, D.; Das, S.; Gastier-Foster, J.; Grody, W.W.; Hegde, M.; Lyon, E.; Spector, E.; 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, 405–424. [Google Scholar] [CrossRef]
  36. Mantelli, M.; Pastorino, L.; Ghiorzo, P.; Barile, M.; Bruno, W.; Gargiulo, S.; Sormani, M.P.; Gliori, S.; Vecchio, S.; Ciotti, P.; et al. Early Onset May Predict G101W CDKN2A Founder Mutation Carrier Status in Ligurian Melanoma Patients. Melanoma Res. 2004, 14, 443–448. [Google Scholar] [CrossRef]
  37. Astiazaran-Symonds, E.; Goldstein, A.M. A Systematic Review of the Prevalence of Germline Pathogenic Variants in Patients with Pancreatic Cancer. J. Gastroenterol. 2021, 56, 713–721. [Google Scholar] [CrossRef]
  38. Wieme, G.; Kral, J.; Rosseel, T.; Zemankova, P.; Parton, B.; Vocka, M.; Van Heetvelde, M.; Kleiblova, P.; Blaumeiser, B.; Soukupova, J.; et al. Prevalence of Germline Pathogenic Variants in Cancer Predisposing Genes in Czech and Belgian Pancreatic Cancer Patients. Cancers 2021, 13, 4430. [Google Scholar] [CrossRef]
  39. Fountzilas, E.; Eliades, A.; Koliou, G.-A.; Achilleos, A.; Loizides, C.; Tsangaras, K.; Pectasides, D.; Sgouros, J.; Papakostas, P.; Rallis, G.; et al. Clinical Significance of Germline Cancer Predisposing Variants in Unselected Patients with Pancreatic Adenocarcinoma. Cancers 2021, 13, E198. [Google Scholar] [CrossRef]
  40. Hu, C.; Hart, S.N.; Polley, E.C.; Gnanaolivu, R.; Shimelis, H.; Lee, K.Y.; Lilyquist, J.; Na, J.; Moore, R.; Antwi, S.O.; et al. Association between Inherited Germline Mutations in Cancer Predisposition Genes and Risk of Pancreatic Cancer. JAMA 2018, 319, 2401–2409. [Google Scholar] [CrossRef]
  41. Cremin, C.; Lee, M.K.-C.; Hong, Q.; Hoeschen, C.; Mackenzie, A.; Dixon, K.; McCullum, M.; Nuk, J.; Kalloger, S.; Karasinska, J.; et al. Burden of Hereditary Cancer Susceptibility in Unselected Patients with Pancreatic Ductal Adenocarcinoma Referred for Germline Screening. Cancer Med. 2020, 9, 4004–4013. [Google Scholar] [CrossRef]
  42. Casolino, R.; Corbo, V.; Beer, P.; Hwang, C.-I.; Paiella, S.; Silvestri, V.; Ottini, L.; Biankin, A.V. Germline Aberrations in Pancreatic Cancer: Implications for Clinical Care. Cancers 2022, 14, 3239. [Google Scholar] [CrossRef]
  43. Ghiorzo, P.; Pastorino, L.; Bonelli, L.; Cusano, R.; Nicora, A.; Zupo, S.; Queirolo, P.; Sertoli, M.; Pugliese, V.; Bianchi-Scarrà, G. INK4/ARF Germline Alterations in Pancreatic Cancer Patients. Ann. Oncol. 2004, 15, 70–78. [Google Scholar] [CrossRef] [PubMed]
  44. Kimura, H.; Klein, A.P.; Hruban, R.H.; Roberts, N.J. The Role of Inherited Pathogenic CDKN2A Variants in Susceptibility to Pancreatic Cancer. Pancreas 2021, 50, 1123–1130. [Google Scholar] [CrossRef] [PubMed]
  45. Shindo, K.; Yu, J.; Suenaga, M.; Fesharakizadeh, S.; Cho, C.; Macgregor-Das, A.; Siddiqui, A.; Witmer, P.D.; Tamura, K.; Song, T.J.; et al. Deleterious Germline Mutations in Patients with Apparently Sporadic Pancreatic Adenocarcinoma. J. Clin. Oncol. 2017, 35, 3382–3390. [Google Scholar] [CrossRef] [PubMed]
  46. McWilliams, R.R.; Wieben, E.D.; Chaffee, K.G.; Antwi, S.O.; Raskin, L.; Olopade, O.I.; Li, D.; Highsmith, W.E.; Colon-Otero, G.; Khanna, L.G.; et al. CDKN2A Germline Rare Coding Variants and Risk of Pancreatic Cancer in Minority Populations. Cancer Epidemiol. Biomark. Prev. 2018, 27, 1364–1370. [Google Scholar] [CrossRef]
  47. Bruno, W.; Dalmasso, B.; Barile, M.; Andreotti, V.; Elefanti, L.; Colombino, M.; Vanni, I.; Allavena, E.; Barbero, F.; Passoni, E.; et al. Predictors of Germline Status for Hereditary Melanoma: 5 Years of Multi-Gene Panel Testing within the Italian Melanoma Intergroup. ESMO Open 2022, 7, 100525. [Google Scholar] [CrossRef]
  48. Pastorino, L.; Andreotti, V.; Dalmasso, B.; Vanni, I.; Ciccarese, G.; Mandalà, M.; Spadola, G.; Pizzichetta, M.A.; Ponti, G.; Tibiletti, M.G.; et al. Insights into Genetic Susceptibility to Melanoma by Gene Panel Testing: Potential Pathogenic Variants in ACD, ATM, BAP1, and POT1. Cancers 2020, 12, E1007. [Google Scholar] [CrossRef]
  49. Dalmasso, B.; Puccini, A.; Catalano, F.; Borea, R.; Iaia, M.L.; Bruno, W.; Fornarini, G.; Sciallero, S.; Rebuzzi, S.E.; Ghiorzo, P. Beyond BRCA: The Emerging Significance of DNA Damage Response and Personalized Treatment in Pancreatic and Prostate Cancer Patients. Int. J. Mol. Sci. 2022, 23, 4709. [Google Scholar] [CrossRef]
  50. Astiazaran-Symonds, E.; Kim, J.; Haley, J.S.; Kim, S.Y.; Rao, H.S.; Genetics Center, R.; Carey, D.J.; Stewart, D.R.; Goldstein, A.M. A Genome-First Approach to Estimate Prevalence of Germline Pathogenic Variants and Risk of Pancreatic Cancer in Select Cancer Susceptibility Genes. Cancers 2022, 14, 3257. [Google Scholar] [CrossRef]
  51. Hannan, Z.; Yu, S.; Domchek, S.; Mamtani, R.; Reiss, K.A. Clinical Characteristics of Patients With Pancreatic Cancer and Pathogenic ATM Alterations. JNCI Cancer Spectr. 2021, 5, pkaa121. [Google Scholar] [CrossRef]
  52. Hosein, A.N.; Dougan, S.K.; Aguirre, A.J.; Maitra, A. Translational Advances in Pancreatic Ductal Adenocarcinoma Therapy. Nat. Cancer 2022, 3, 272–286. [Google Scholar] [CrossRef]
  53. Javle, M.; Shacham-Shmueli, E.; Xiao, L.; Varadhachary, G.; Halpern, N.; Fogelman, D.; Boursi, B.; Uruba, S.; Margalit, O.; Wolff, R.A.; et al. Olaparib Monotherapy for Previously Treated Pancreatic Cancer with DNA Damage Repair Genetic Alterations Other Than Germline BRCA Variants: Findings From 2 Phase 2 Nonrandomized Clinical Trials. JAMA Oncol. 2021, 7, 693–699. [Google Scholar] [CrossRef]
  54. Maio, M.; Ascierto, P.A.; Manzyuk, L.; Motola-Kuba, D.; Penel, N.; Cassier, P.A.; Bariani, G.M.; De Jesus Acosta, A.; Doi, T.; Longo, F.; et al. Pembrolizumab in Microsatellite Instability High or Mismatch Repair Deficient Cancers: Updated Analysis from the Phase 2 KEYNOTE-158 Study. Ann. Oncol. 2022, in press. [Google Scholar] [CrossRef]
  55. Aslanian, H.R.; Lee, J.H.; Canto, M.I. AGA Clinical Practice Update on Pancreas Cancer Screening in High-Risk Individuals: Expert Review. Gastroenterology 2020, 159, 358–362. [Google Scholar] [CrossRef]
  56. Canto, M.I.; Almario, J.A.; Schulick, R.D.; Yeo, C.J.; Klein, A.; Blackford, A.; Shin, E.J.; Sanyal, A.; Yenokyan, G.; Lennon, A.M.; et al. Risk of Neoplastic Progression in Individuals at High Risk for Pancreatic Cancer Undergoing Long-Term Surveillance. Gastroenterology 2018, 155, 740–751.e2. [Google Scholar] [CrossRef]
  57. Paiella, S.; Salvia, R.; De Pastena, M.; Pollini, T.; Casetti, L.; Landoni, L.; Esposito, A.; Marchegiani, G.; Malleo, G.; De Marchi, G.; et al. Screening/Surveillance Programs for Pancreatic Cancer in Familial High-Risk Individuals: A Systematic Review and Proportion Meta-Analysis of Screening Results. Pancreatology 2018, 18, 420–428. [Google Scholar] [CrossRef]
  58. Signoretti, M.; Bruno, M.J.; Zerboni, G.; Poley, J.-W.; Delle Fave, G.; Capurso, G. Results of Surveillance in Individuals at High-Risk of Pancreatic Cancer: A Systematic Review and Meta-Analysis. United Eur. Gastroenterol. J. 2018, 6, 489–499. [Google Scholar] [CrossRef]
  59. Paiella, S.; Capurso, G.; Cavestro, G.M.; Butturini, G.; Pezzilli, R.; Salvia, R.; Signoretti, M.; Crippa, S.; Carrara, S.; Frigerio, I.; et al. Results of First-Round of Surveillance in Individuals at High-Risk of Pancreatic Cancer from the AISP (Italian Association for the Study of the Pancreas) Registry. Am. J. Gastroenterol. 2019, 114, 665–670. [Google Scholar] [CrossRef]
  60. Capurso, G.; Paiella, S.; Carrara, S.; Butturini, G.; Secchettin, E.; Frulloni, L.; Zerbi, A.; Falconi, M. Italian Registry of Families at Risk of Pancreatic Cancer: AISP Familial Pancreatic Cancer Study Group. Dig. Liver Dis. 2020, 52, 1126–1130. [Google Scholar] [CrossRef]
Figure 1. Comparison between the relative frequency of candidate susceptibility genes PVs in our cohort and literature data according to the systematic review by Astiazaran-Symonds E and Goldstein AM [37].
Figure 1. Comparison between the relative frequency of candidate susceptibility genes PVs in our cohort and literature data according to the systematic review by Astiazaran-Symonds E and Goldstein AM [37].
Cancers 14 04447 g001
Table 1. Characteristics of the study cohort according to the germline status.
Table 1. Characteristics of the study cohort according to the germline status.
Overall (N = 422)Non-Carriers (N = 352)PV Carriers (N = 70)p-Value
Age, mean (range)67 (30–92)67.4 (30–92)64.3 (44–82)0.0254
Female, N (%)221 (52)189 (54)32 (46)0.222
Initial stage, N (%)
Resectable92 (22)76 (22)16 (23)0.323
Borderline25 (6)21 (6)4 (6)
Unresectable51 (12)38 (11)13 (19)
Metastatic188 (45)158 (45)30 (43)
Missing66 (16)59 (17)7 (10)
FH for PC, N (%)55 (13)37 (11)18 (26)0.001
Personal/FH for BC/Ovarian, N (%)76 (18)51 (14)25 (36)<0.001
Personal/FH for melanoma, N (%)30 (7)17 (5)13 (19)<0.001
Other primary cancers, N (%)64 (16)43 (13)21 (32)<0.001
Pts = patients; wt = wild-type; PV = pathogenic variant/likely pathogenic variant; FH = family history. PC = pancreatic cancer; BC = breast cancer; significant values are shown in bold.
Table 2. Pathogenic variant frequency: overall and according to personal and family history.
Table 2. Pathogenic variant frequency: overall and according to personal and family history.
GenesCumulative
Frequency
FH of PCPersonal and/or FH of BC and OvaryPersonal and/or FH of Melanoma Frequency in
Sporadic PC
n = 422n = 55n = 76n = 30n = 283
n%n%n%n%n%
ATM9(2.1)3(5.4)1(1.3)1(3.3)5(1.7)*
BRCA16(1.4)1(1.8)5(6.6)1(3.3)--**
BRCA213(3)1(1.8)7(9.2)2(6.7)5(1.7)*/**
BRCA1/219(4.5)2(3.6)12(15.8)3(10)5(1.7)*/**
CDKN2A19(4.5)8(14.5)5(6.6)6(20)8(2.8)*/**
CHEK27(1.6)1(1.8)--1(3.3)5(1.7)
COL7A15(1.2)1(1.8)1(1.3)--3(1)
ERCC41(0.2)------1(0.3)
FANCA/C/G3(0.7)2(3.6)2(2.6)1(3.3)--**
MLH11(0.2)--1(1,3)----
MSH22(0.4)--1(1.3)--1(0.3)
NBN3(0.7)--1(1.3)1(3.3)2(0.7)*
RAD50/512(0.4)1(1.8)1(1.3)1(3.3)- -**
RET11(0.2)------1(0.3)
SDHA1(0.2)------1(0.3)
TP531(0.2)--1(1.3)--
All genes74(17.5)18(32.7)26(34.2)14(46.7)32(11.3)
FH = family history; PC = pancreatic cancer; BC = breast cancer. * 2 or more variants in the same patient; ** some patients were included in more than one category.
Table 3. Cox proportional hazards models to identify characteristics associated with prognosis in terms of overall survival.
Table 3. Cox proportional hazards models to identify characteristics associated with prognosis in terms of overall survival.
Univariable Cox ModelsMultivariable Cox Model N = 340
HR (95% CI)p-ValueHR (95% CI)p-Value
Age (10-years) ***1.18 (1.03–1.32)0.0041.18 (1.06; 1.33)0.004
Male vs. Female *1.06 (0.85–1.31)0.6121.13 (0.91; 1.40)0.270
Initial stage (N = 353) **
Resectable1.00Ref1.00Ref
Borderline1.24 (0.66–2.30)0.5041.25 (0.66; 2.39)0.491
Unresectable1.39 (0.93–2.08) 0.1131.39 (0.92; 2.09)0.116
Metastatic3.20 (2.38–4.29)<0.0013.39 (2.51; 4.57)<0.001
FH for PC *0.85 (0.62–1.17)0.3150.90 (0.65; 1.24)0.526
Personal/FH for BC/Ovarian *0.91 (0.69–1.21)0.5190.92 (0.69; 1.22)0.552
Personal/FH for melanoma *0.99 (0.66–1.49)0.9691.08 (0.71; 1.63)0.723
Other primary cancers, (N = 395) *0.76 (0.56–1.04)0.0890.74 (0.54; 1.01)0.061
Presence of any PV vs. WT *0.78 (0.59–1.04)0.0900.81 (0.61; 1.09)0.160
Multivariable estimates are adjusted for * age, ** age, sex and PV, *** sex. FH = family history; WT = wild-type; PV = Pathogenic Variant/Likely Pathogenic Variant; PC = pancreatic cancer; BC = breast cancer; significant values are shown in bold.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Puccini, A.; Ponzano, M.; Dalmasso, B.; Vanni, I.; Gandini, A.; Puglisi, S.; Borea, R.; Cremante, M.; Bruno, W.; Andreotti, V.; et al. Clinical Significance of Germline Pathogenic Variants among 51 Cancer Predisposition Genes in an Unselected Cohort of Italian Pancreatic Cancer Patients. Cancers 2022, 14, 4447. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14184447

AMA Style

Puccini A, Ponzano M, Dalmasso B, Vanni I, Gandini A, Puglisi S, Borea R, Cremante M, Bruno W, Andreotti V, et al. Clinical Significance of Germline Pathogenic Variants among 51 Cancer Predisposition Genes in an Unselected Cohort of Italian Pancreatic Cancer Patients. Cancers. 2022; 14(18):4447. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14184447

Chicago/Turabian Style

Puccini, Alberto, Marta Ponzano, Bruna Dalmasso, Irene Vanni, Annalice Gandini, Silvia Puglisi, Roberto Borea, Malvina Cremante, William Bruno, Virginia Andreotti, and et al. 2022. "Clinical Significance of Germline Pathogenic Variants among 51 Cancer Predisposition Genes in an Unselected Cohort of Italian Pancreatic Cancer Patients" Cancers 14, no. 18: 4447. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14184447

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop