Skip to main content
Erschienen in: BMC Gastroenterology 1/2018

Open Access 01.12.2018 | Research article

Demographic, clinical, and pathological features of early onset pancreatic cancer patients

verfasst von: Chara Ntala, Silvana Debernardi, Roger M. Feakins, Tatjana Crnogorac-Jurcevic

Erschienen in: BMC Gastroenterology | Ausgabe 1/2018

Abstract

Background

Early onset pancreatic cancer (EOPC), i.e. pancreatic ductal adenocarcinoma (PDAC) occurring in patients below 50 years of age, is rare and there is limited information regarding risk factors, molecular basis and outcome. This study aimed to determine the demographic and clinicopathological features and survival figures for EOPC.

Methods

A retrospective analysis of patients treated at the Royal London Hospital for PDAC between September 2004 and September 2015 was performed. Data on demographics, risk factors, presentation, pathological features, treatment and survival outcome were compared in EOPC and older PDAC patients.

Results

Of 369 PDAC cases identified, 35 (9.5%) were EOPC. Compared to older patients, EOPC patients were more frequently male (71% vs 54%, p = 0.043) and less commonly of British origin (37% vs 70%, p = 0.002). There was no significant difference regarding the prevalence of any of the risk factors known to be associated with older PDAC patients. Fewer EOPC patients presented with resectable disease (23% vs 44%, p = 0.015) and more received adjuvant chemo/radiotherapy (60% vs 46%, p = 0.008). The overall median survival and stage specific survival did not differ significantly between the two groups, although a longer survival for localized disease was seen in EOPC patients (25 months (12.9–37, 95%CI) vs 13 months (10.5–15.5 95%CI) for older PDAC patients).

Conclusions

The EOPC patients had different demographics and were more likely than their older PDAC counterparts to be male. Typically they presented with more advanced disease, received more aggressive treatment, and had on overall similar survival outcome.
Abkürzungen
AJCC
American Joint Committee on Cancer
BMI
Body mass index
EOPC
Early onset pancreatic cancer
FNA
Fine needle aspiration
HP
Hereditary pancreatitis
HPB
Hepato-Pancreato-Biliary
PDAC
Pancreatic ductal adenocarinoma
PJS
Peutz Jeghers syndrome
TNM
Tumour node metastasis
UICC
Union for International Cancer Control
WHO
World health organisation

Background

Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy. It is the fourth leading cause of cancer death with a 7% five-year survival rate in the United States [1], and is predicted to be the second cause of cancer-related death by 2030 [2]. The mean age of PDAC patients at presentation is 71 years [3]; however, 5–10% of these patients are diagnosed with this malignancy at a young age, when they are less than 50 years old [47]. This important subgroup of PDAC patients, often referred to as early onset pancreatic cancer (EOPC), has been poorly studied, and the cause of such an early presentation of the disease remains unknown [6, 814].
PDAC is considered to be more frequent in individuals with familial history [1518] and hereditary genetic syndromes, such as Hereditary Pancreatitis (HP) [1921], Lynch syndrome [22] and Peutz-Jeghers syndrome [23, 24]. Modifiable risk factors include tobacco exposure, alcohol use, chronic pancreatitis, diabetes mellitus, diet, obesity, previous radiotherapy, as well as certain types of abdominal surgery and infections [2533]. Also, an inverse association between PDAC and atopic diseases has been identified [34].
In this report, we provide the data of a comprehensive and systematic study on demographics and known PDAC risk factors, as well as detailed clinicopathological, treatment and outcome figures for EOPC patients. The study comprised retrospective examination of notes on PDAC patients collected over the 11-year period between September 2004 and September 2015, from one of the major Hepato-Pancreato-Biliary (HPB) referral hospital centres in London, UK. We compared these data to the data collected for the older PDAC patients from the same centre in order to identify differences and potentially suggest clues to the biology of the disease.

Methods

Database search and data extraction

We conducted a search of the pathology archives and patients’ database at the Royal London Hospital for period between 18/9/2004 to 18/9/2015 in order to identify patients who were diagnosed with pancreatic ductal adenocarcinoma. We used the topography search codes “pancreatic structure” and “pancreas and duodenum”, and included pancreatic FNAs (fine needle aspirations), core biopsies and surgical resection specimens with a pathology diagnosis of pancreatic ductal adenocarcinoma or adenocarcinoma with features “compatible with”, “suggestive of” or “typical of” pancreatic ductal origin. We excluded all other histologic subtypes and cases of adenocarcinoma of unknown origin, highly suspicious of adenocarcinoma, adenocarcinoma arising from ampulla or bile duct and non-neoplastic diagnoses (Fig. 1). Patients under the age of 50 were grouped into the EOPC cohort and were compared to older patients, referred to here as the older PDAC group. Information extracted consisted of the following: 1) Demographics: age, gender and ethnicity. 2) Epidemiologic and genetic information: history of alcohol excess, obesity (BMI > 30), previous abdominal surgery or radiotherapy, allergy status, and family history of pancreatic cancer or other related malignancies. Specific quantitative data on smoking history in terms of duration, intensity, or recency were rarely available thus smoking history was descriptive (never/ex/current smoker) rather than in accordance with specific definitions (e.g. pack-years). 3) Presenting symptoms: Past medical history of chronic pancreatitis was considered positive if it was present more than 6 months before diagnosis, and recent onset diabetes mellitus was defined as diabetes diagnosed within 3 years before diagnosis. 4) Clinical stage at diagnosis: record followed the American Joint Committee on Cancer (AJCC) and the Union for International Cancer Control (UICC) TNM staging system 7th edition [35]. 5) Surgical management, details of adjuvant and palliative treatment. 6) Survival data. The date of diagnosis was defined as the date when biopsy was taken. The date of last follow-up was used if date of death was not available. The study was done under Research Ethics approval (Reference Number: 05/Q0408/65).

Statistical analysis

Differences between groups were calculated with two-sided chi-square test (p < 0.05 was considered statistically significant). The age was presented as a mean value with ranges, and survival was presented as a median value with 95% confidence intervals (CI). Survival curves were estimated using the Kaplan–Meier method and compared by the log-rank test. All statistical analyses were performed using IBM SPSS Statistics 24.0.

Results

Patient demographics, epidemiologic and genetic characteristics

Our search identified 1275 cases; of 369 pancreatic cancer cases 35 were EOPC, comprising 9.5% of the total PDAC population (Fig. 1). The general characteristics of EOPC and older PDAC patients are presented in Table 1. The mean age was 45.71 (33 to 50) years for the young onset population while older PDAC patients had a mean age of 66.19 (51 to 85) years. Gender distribution was significantly different between EOPC and older PDAC groups, with higher rate of male patients in the younger cohort (71% vs 54%, p = 0.043). In terms of ethnicity, the majority of older PDAC patients were British (70% vs 37% in EOPC cohort, p = 0.002), whereas the EOPC group included a significant proportion of patients from Asian and Central/Eastern European origin. There was no significant difference in terms of modifiable and genetic risk factors between the two groups. First-degree family history in the EOPC cohort is presented in Table 2. Only one of the 35 EOPC patients was affected by a genetic syndrome (Lynch syndrome). Four EOPC (11%) patients and 34 older PDAC patients (10%) had a first-degree relative with history of any type of cancer (p = 0.989). None of the EOPC and only 12 (4%) of the older PDAC patients had a first-degree relative with pancreatic cancer (p = 0.25). Interestingly, one EOPC patient had two 2nd degree relatives from parental side (an uncle and a cousin) also diagnosed with pancreatic cancer at an early age (50 and 49 years, respectively).
Table 1
EOPC and older PDAC demographics, past medical history, environmental and genetic risk factors
Characteristics
EOPC (35), % (≤50 years)
PDAC (334), % (> 50 years)
p value
Mean Age
45.71
66.19
 
(Range in years)
(33 to 50)
(51 to 85)
 
Gender
  
0.043
 Male
25 (71%)
179 (54%)
 
 Female
10 (29%)
155 (46%)
 
Ethnicity
  
0.002
 British
13 (37%)
233 (70%)
 
 Asian
5 (14%)
9 (3%)
 
 Black
3 (9%)
22 (7%)
 
 Other white background
4 (11%)
11 (3%)
 
 Any other group
2 (6%)
12 (4%)
 
 Missing
8 (23%)
47 (14%)
 
Smoking status
  
0.850
 Current smoker
7 (20%)
55 (16%)
 
 Ex-smoker
4 (11%)
45 (13%)
 
 No
13 (37%)
109 (33%)
 
 Missing
11 (31%)
125 (37%)
 
History of alcohol excess
  
0.556
 Yes
9 (26%)
69 (21%)
 
 No
14 (40%)
140 (42%)
 
 Missing
12 (34%)
125 (37%)
 
Obesity
  
0.244
 Yes
2 (6%)
23 (7%)
 
 No
8 (23%)
37 (11%)
 
 Missing
25 (71%)
274 (82%)
 
Genetic syndrome
  
0.989
 Yes
1 (3%)
9 (3%)
 
 No
30 (86%)
266 (80%)
 
 Missing
4 (11%)
59 (18%)
 
History of chronic pancreatitis
  
0.092
 Yes
5 (14%)
96 (29%)
 
 No
23 (66%)
191 (57%)
 
 Missing
7 (20%)
47 (14%)
 
History of diabetes
  
0.385
 Yes
8 (23%)
58 (17%)
 
 No
22 (63%)
233 (70%)
 
 Missing
5 (14%)
43 (13%)
 
Allergya
  
0.853
 Yes
2 (6%)
35 (10%)
 
 No
15 (43%)
228 (68%)
 
 Missing
18 (51%)
71 (21%)
 
Previous radiotherapy
  
0.838
 Yes
1 (3%)
14 (4%)
 
 No
23 (66%)
261 (78%)
 
 Missing
11 (31%)
59 (18%)
 
Previous abdominal surgeryb
  
0.056
 Yes
9 (26%)
78 (23%)
 
 No
9 (26%)
194 (58%)
 
 Missing
17 (49%)
62 (19%)
 
aAllergy to penicillin; bPrevious abdominal surgery included cholecystectomy, lateral pancreaticojejunostomy
Table 2
Family history of cancer in EOPC patients
EOPC patients
Family history of cancer
1
Mother: endometrial cancer
2
Mother: colon and endometrial cancer,
mAunt: endometrial cancer, pGrandmother: bowel cancer, pCousin: breast cancer (age 32), Grandmother: stomach cancera
3
Father: esophageal cancer, Brother: colon cancer, Sister: esophageal cancer
4
Sister: lung cancer (age 38)
p: paternal, m: maternal, aLynch syndrome

Symptoms at presentation

With regards to symptoms at presentation, obstructive jaundice was the most frequent presenting symptom in both groups, followed by anorexia, abdominal pain, nausea and vomiting. Two EOPC (6%) patients presented with early onset diabetes compared to 40 PDAC (12%) patients (p = 0.299).

Pathology and clinical stage at diagnosis

A significantly lower number of EOPC patients (22.9%) presented with localised resectable disease compared to older PDAC patients (44%) (p = 0.015). A similar proportion of EOPC (54%) and older PDAC (42%) patients had locally advanced unresectable disease or presented with distant metastases (23% vs 17%). In EOPC, half of the patients with metastases at diagnosis had a single liver metastasis and the other half had more than one site of metastasis. The most common sites of metastatic disease were liver (75%), lung (37.5%), pleura and omentum (12.5% each). Similarly, in older PDAC patients the most common site of metastasis was liver, occurring alone in just above half of the cases (53.5%) and in combination with other sites in a further 15.5% of tumours. Other single sites were lung (7%), peritoneum (7%), omentum (1.7%) and pleura (1.7%). Tumour grading and AJCC/UICC TNM stage of EOPC and older PDAC patients did not differ, and there was no significant difference in terms of the other pathological characteristics of tumours (location, differentiation, stage, perineural invasion and vascular invasion) between the two groups (Table 3).
Table 3
EOPC and older PDAC tumour characteristics and treatment details
Tumour Characteristics
EOPC (N = 35), % (≤50 years)
PDAC (N = 334), % (> 50 years)
p value
Resectable
  
0.015
 Yes
8 (22.9%)
147 (44%)
 
 No
27 (77.1%)
186 (56%)
 
 Missing
0
1 (0%)
 
Location
  
0.579
 Head/Uncinate
27 (77%)
265 (79%)
 
 Body/Tail
8 (23%)
62 (19%)
 
 Missing
0
7 (2%)
 
Differentiation grade
  
0.315
 Well
2 (6%)
15 (4%)
 
 Moderate
16 (46%)
127 (38%)
 
 Poor
12 (34%)
170 (51%)
 
 Missing
5 (14%)
22 (7%)
 
Stages
  
0.194
 I
2 (5.8%)
16 (5%)
 
 II*
6 (17.1%)
117(34%)
 
 III
19 (54.2%)
140 (42%)
 
 IV
8 (22.9%)
57 (17%)
 
 Missing
0
4 (1%)
 
Perineural invasion
  
0.348
 Yes
12 (34%)
142 (43%)
 
 No
23 (66%)
192 (57%)
 
Perivascular invasion
 Yes
7 (20%)
122 (37%)
0.051
 No
28 (80%)
212 (63%)
 
Chemotherapy/ radiotherapy
  
0.008
 Yes
21 (60%)
153 (46%)
 
 No
5 (14%)
131 (39%)
 
 Missing
9 (26%)
50 (15%)
 
*Stage II p value = 0.032

Treatment and survival

All 8 (22.9%) EOPC and 147 (44%) PDAC patients with localised disease underwent resection with curative intent. The AJCC/UICC TNM stage of EOPC patients who underwent resection was as follows: IA (n = 2), IIA (n = 1), IIB (n = 5). Sixty per cent of EOPC patients received adjuvant or palliative chemotherapy/radiotherapy whereas only 46% of the older PDAC patients were fit enough to receive the same treatment (p = 0.008) (Table 3). Six (17.4%) EOPC and 77 (23%) PDAC patients received a combination of chemotherapy/radiotherapy and surgery with curative intent, out of which one (16.7%) EOPC and six (7.8%) PDAC received this as neoadjuvant treatment that down-staged their disease. The median overall survival of the EOPC patients (12 months, 5–18.9, 95% CI) was higher than in older PDAC patients (9 months, 7.8–10.2, 95% CI) and there was also a trend towards increased stage specific survival (25 vs to 13 months) but neither difference was statistically significant (Fig. 2 and Table 4). Similarly, for patients with locally advanced disease, the median overall survival was 11 months (3.9–18.1, 95%CI) for the EOPC patients and 8 months (6.5–9.4, 95%CI) for the older PDAC patients (p = 0.172). Regarding patients with metastatic disease, both cohorts had a median survival of 6 months (p = 0.213) (Table 4).
Table 4
Overall survival (in months) of EOPC and older PDAC cohorts
 
Median survival (months)
 
EOPC (N = 35) (≤50 years)
PDAC (N = 334) (> 50 years)
p value
Entire cohorta
12 (5–18.9 95%CI)
9 (7.8–10.2 95%CI)
0.168
Stage I-II
25 (12.9–37 95%CI)
13(10.5–15.5 95%CI)
0.307
Stage III
11 (3.9–18.1 95%CI)
8 (6.5–9.4 95%CI)
0.172
Stage IV
6 (3.2–8.8 95%CI)
6 (4.8–7.2 95%CI)
0.213
aData missing for three EOPC and nine PDAC patients

Discussion

In this study, we performed a detailed retrospective analysis of EOPC patients with histologically confirmed PDAC in the setting of one of the largest HPB centres in the United Kingdom. While previous EOPC reports also included patients with pathological “variants” (e.g. mucinous cystic neoplasms) which are not included in the current WHO classification of pancreatic ductal adenocarcinoma, our report is based on the most recent PDAC classification and staging. We have used 50 years as the cut-off age for EOPC patients, which, although arbitrary, has been used previously [47]. A detailed summary of comparisons between different EOPC studies is shown in Table 5. The main differences between the two PDAC groups were seen in demographic data. EOPC patients in our population were more frequently males, in agreement with previous studies [57, 12, 13, 36, 37]. In addition, our EOPC patients were less frequently of white British background and more commonly Asian or from ‘any other white background’. The latter comprised mainly Central and Eastern Europeans. Similar findings were reported by Raimondi et al. who demonstrated in a worldwide study a higher number of male than female patients and a peak in Central and Eastern European countries followed very closely by Asian countries [28]. The association of gender and race/ethnic group with incidence of pancreatic cancer has also been documented in a US report conducted by the National Cancer Institute, where the authors showed the higher incidence and mortality rate in men than in women in each racial/ethnic group, between the age of 30 and 54 [38]. Miller et al., showed up to 50% higher incidence and mortality rate in the black compared to the white population, for the same age group (30–54 years) [38].
Table 5
Comprehensive comparison of 14 studies on EOPC, including our study (first column)
 
Ntala C 2018, RLH, UK
Piciucchi M 2016, Italy [7]
Tingstedt B 2011, Sweden [5]
Luttges J 2003, Germany [8]
Bergmann F 2006, Germany [9]
Duffy A 2009, USA [10]
He J 2013, USA [12]
Length of the study in years
11y (2004–2015)
7y (2006–2013)
15y (1993–2008)
26y (1973–1999)
2006
13y (1995–2008)
34y (1975–2009)
DEMOGRAPHIC
 No. EOPC (%)
35 (9.5%)
25 (8.5%)
33 (5.7%)
10 (2.3%)
7 cases
136 (4.4%)
75 (7.9%)
 Age included in study (range)
≤50y (33–50)
≤50y
≤50y (30–50)
≤39 (10-39y)
≤40y (35–40)
≤45y (20–45)
≤45y (31–45)
 Male
71% (p = 0.043)
68.0%
61.0% (p = 0.02)
100.0%
0
54.0%
56.0%
 White
37% (p = 0.002)
     
89.0%
 Black
9.0%
     
4.0%
 Other
31.0%
     
7.0%
RISK FACTORS
 Smokers
31.0%
56%* (p = 0.001)
73.0%
 
71.4%
37.0%
37.0%
 Use of alcohol
26.0%
36.0%
21.0%
    
 Obesity (BMI > 30)
6.0%
mean BMI = 27
9.0%
    
 History of CP
14.0%
0.0%
12.0%
 
14.3%
13.0%
 
 History of diabetes
23.0%
4.0% (p = 0.00001)
3.0%
    
 Family history of cancer (1st degree relative)
11.4%
48.0%
  
71.4%
5.1%
 
 Family history of pancreatic cancer (1st degree relative)
0.0%
8.0%
3.0%
 
0
2.2%
 
SYMPTOMS AT PRESENTATION
 New onset diabetes
6.0%
4.0%
21.0%
    
 Weight loss
54.3%
52.0%
55.0%
   
33.0%
 Jaundice
62.8%
16.0% (p = 0.006)
61.0%
   
45.0%
 Abdominal pain
45.7%
68.0% (p = 0.06)
91.0% (p = 0.001^)
   
32.0% (p = 0.06)
PATHOLOGY
 Location-Head
77.0%
64.0% (p = 0.03)
79.0%
 
71.40%
  
 Location-Body
(B + T) 23%
 
12.0%
    
 Location-Tail
  
9.0%
 
28.60%
  
 G1 - Well differentiated
5.7%
 
20.0%
10.0%
57.2%
  
 G2 - Moderately differentiated
45.7%
 
45.0%
45%
42.8%
  
 G3 - Poorly differentiated
34.3%
61.0%
25.0%
14%
   
 Perineural invasion
34.3%
     
64.0%
 Localised/Resectable
22.9% (p = 0.015)
16.0%
18.0%
  
25.7%
32.0%
 Locally advanced
54.0%
36.0%
27.0% (p = 0.005)
  
20.1%
68.0%
 Metastatic
22.9%
48.0%
52.0% (p = 0.001)
 
14.3%
50.0%
 
TREATMENT
 Resected cases
22.9% (p = 0.015)
16.0%
27.0% (p = 0.01)
 
71.4%
25.7%
(R0) 69.0%
 Palliative chemo/radiotherapy
60.0% (p = 0.008)
48.0% (p = 0.003)
45.0%/36.0% (p = ns/0.002)
 
28.6%
  
 Supportive treatment
17.1%
      
SURVIVAL (months)
 Median OS
12
11
5.7
  
12.3
19
 Median OS (resectable cases)
25
    
41.8
 
Stage associated survival (%; (OS, months))
 Stage I + II
22.8% (25)
    
25.7% (41.8) (p = 0.0001)
I + IIA:32% (27)
 Stage III
54.3% (11)
36.0% (11)
   
20% (15.3)
IIB + III;68% (16)
 Stage IV
17.1% (6)
48.0% (7)
   
50% (7.2)
 
Survival rate
 1 year survival rate
 5 years survival rate
     
3.3%
24.00%
 10 years survival rate
      
17.00%
 5 years survival rate I-IIA
      
42.00%
 5 years survival rate IIB-III
      
16.00%
 
Beeghly-Fadiel A 2016, USA [6]
McWilliams RR 2017, USA [14]
Ohmoto A 2017, Japan [37]
Jiang Q-L 2017, China [13]
Lin J-C 2011, China [4]
Soliman AS 2002, Egypt [41]
Raissouni S 2012, Morocco [36]
Length of the study in years
25y (1988–2013)
11y (2000–2011)
11y (2002–2013)
16y (1999–2014)
19y (1990–2009)
5y (1995–2000)
5y (2005–2010)
DEMOGRAPHIC
 No. EOPC (%)
118 (8.4%)
226 (11.5%)
17 (1.87%)
156 (8.7%)
25 (10.2%)
165 (22.6%)
32 (17%)
 Age included in study (range)
≤49y
≤44y
≤40y (21–40)
≤45y (17–45)
≤49y
≤50y
≤45y (28–45)
 Male
60.2%
54.0%
64.7%
75% (p = 0.006)
  
65.0%
 White
86.3%
      
 Black
11.10%
      
 Other
2.60%
      
RISK FACTORS
 Smokers
 
35% and 6%§
58.8%
34.6% (p = 0.024)
44.0%
 
12.5%
 Use of alcohol
 
11%†
 
35.3%
32.0%
  
 Obesity (BMI > 30)
 
19%
 
(BMI ≥ 28) 16.7%
   
 History of CP
 
1.0%
  
16.0%
  
 History of diabetes
 
3.0%
 
11.5%
4.0%
 
6.0%
 Family history of cancer (1st degree relative)
  
29.4%
17.3%
   
 Family history of pancreatic cancer (1st degree relative)
17.5%
8.0%
5.8%
3.8%
8.0%
  
SYMPTOMS AT PRESENTATION
 New onset diabetes
   
11.5%
4.0%
  
 Weight loss
   
46.2%
48.0%
 
43.0%
 Jaundice
   
18.9%
52.0%
 
68.0%
 Abdominal pain
   
62.1%
72.0%
 
87.5%
PATHOLOGY
 Location-Head
67.0%
 
52.90%
69.90%
  
75.0%
 Location-Body
12.3%
 
(B + T) 47.1%
(B + T) 30%
  
12.5%
 Location-Tail
12.3%
     
12.5%
 G1 - Well differentiated
   
3.8%%
   
 G2 - Moderately differentiated
   
14.7%%
   
 G3 - Poorly differentiated
   
26.3%%
   
 Perineural invasion
       
 Localised/Resectable
40.0%
  
28.8%
40.0%
 
18.7%
 Locally advanced
16.0%
  
34.0%
  
21.8%
 Metastatic
44.0%
  
37.2%
52.0%
 
59.3%
TREATMENT
 Resected cases
23.3%
 
23.20%
20.5%
40.0%
33.1%
18.7%
 Palliative chemo/radiotherapy
38.9%
 
70%
47.4%
 
30.7%
26.2%
 Supportive treatment
37.9%
    
14.6%
37.5%
SURVIVAL (months)
 Median OS
9.3 (p = 0.045)
 
6.7
8
5.6
 
6.6
 Median OS in resectable
  
19.6
19
10.3
 
32
Stage associated survival (%; (OS, months))
 Stage I + II
6% + 34%
 
23.2% (19.6)
9% (19)
  
18.8% (32)
 Stage III
16.0%
 
23.5% (18.2)
53.8% (9)
  
21.8% (7.9)
 Stage IV
44.0%
 
52.9% (5)
37.2% (5)
  
59% (6.4)
Survival rate
 1 year survival rate
  
37.5%
 
28% (40% in resected cases)
  
 5 years survival rate
    
4% (10% in resected cases)
  
 10 years survival rate
 5 years survival rate I-IIA
 5 years survival rate IIB-III
*Smoking starting age 19.8y (p value:0.001); ^ value for matched controls; § % for ≤19 and ≥ 40 packets/year; † > 26 g; Cp = chronic pancreatitis; B + T = body + tail; y = years; p = p-value referring to comparison with the older PDAC population (where data available)
One of the putative explanations for the higher incidence of EOPC in males is smoking, a known independent risk factor for pancreatic cancer in all age groups [3, 7, 28, 30, 39, 40]. Indirect association of EOPC and smoking in males was first highlighted by Raimondi et al. who correlated the higher EOPC male/female ratio positively with an early onset of lung cancer (< 50 years of age) [28]. Direct association was provided by Piciucchi et al. [7], who, based on patient interviews, demonstrated a significantly increased risk of EOPC among the ‘current smokers’ group and a positive correlation with the ‘young age at smoking initiation’ [7]. In our cohort, however, only 31% of the total EOPC population had a smoking history which was comparable to the older PDAC cases (29%).
Interestingly, the highest incidence of EOPC patients was reported in two independent studies conducted in North Africa, in Morocco [36] and Egypt [41]: 17% of PDAC patients were younger than 45 years in Morocco [36], and almost 25% in the East Nile Delta were under the age of 50 [41]. In both countries the male to female ratio was around 2:1, in accordance with other reports. Regarding smoking, in the Moroccan population only 12.5% of all EOPC patients were smokers [36]. In contrast, in Egypt smoking is highly prevalent (40% of the general population are smokers). While the authors do not report the smoking history of the EOPC patients, they speculate that occupational and environmental exposure to heavy metals like cadmium, nickel and chromium, as well as other polluting chemicals, could contribute to the high incidence of EOPC in this heavily industrialised region [41]. Unfortunately, we do not have any information on environmental exposure of our study population, and the effect of pollution on our London-based patients would be interesting to explore.
Our study did not identify any differences between EOPC and older PDAC patients in any of the previously identified risk factors for PDAC, i.e. alcohol intake, obesity, history of chronic pancreatitis, history of diabetes, previous abdominal surgery and previous radiotherapy. The rate of recent onset diabetes was somewhat lower in EOPC (6% EOPC vs 12% PDAC, p = 0.299) although this was not statistically significant.
None of the patients in our EOPC cohort met the criteria for a familial pancreatic cancer syndrome [42]. Four of the EOPC patients in our cohort had a family history of any cancer but none had a first-degree relative with pancreatic cancer, and only one patient had a hereditary genetic syndrome (Lynch Syndrome) associated with increased risk of pancreatic cancer [22]. A similarly low incidence of familial cases, with no significant difference between young and old PDAC groups, has been reported in other studies. In the cohort described by Duffy et al. [10], only 2.2% of EOPC patients had a family history of pancreatic cancer and no EOPC patients were affected by any of the hereditary syndromes. Tingstedt et al. [5] reported 3% of EOPC with a first-degree relative with pancreatic cancer. However, somewhat higher incidences were found by Piciucchi et al. [7] in both young and older patients, where 8% and 6.3% of cases respectively had a family history of pancreatic cancer. A recent study by Ohmoto et al. [37] suggests a lack of association between hereditary genetic factors and EOPC. The authors assessed the mutation status of 49 genes involved in hereditary syndromes in the germline of EOPC patients younger than 40 years, but did not find any variants [37]. In contrast, James et al. [17] reported an overall incidence of familial pancreatic cancer of 3%, with the percentage of patients ≤50 years of age being significantly higher than among the sporadic cancer patients (36% and 18.3%, respectively, p = 0.017). Overall, the underlying factors influencing the young onset of pancreatic cancers remain to be determined.
In our study, the presenting symptoms (obstructive jaundice, abdominal pain, and change in bowel habit, nausea /vomiting, anorexia or weight loss) were largely shared between the two cohorts, and both young and old PDAC patients most commonly presented with obstructive jaundice, probably due to a high incidence of lesions located in the pancreatic head. Interestingly, in a study by Piciucchi et al. [7], jaundice at presentation occurred in only 16% of the EOPC patients, a significantly lower rate than in older PDAC patients (44%, p = 0.06) [7]. This was probably due to a lower rate of tumours located in the head of the pancreas (64% vs 83%, p = 0.03). Jiang et al. [13] also reported a low frequency of jaundice in EOPC patients, although this could not be explained by tumour location.
The two groups also showed similar pathological characteristics. Poorly differentiated tumours tended to be more common in older PDAC patients (34% vs 51%) and moderately differentiated in EOPC (46% vs 38%), but this was not statistically significant. Furthermore, no difference was observed in the rates of perineural and vascular invasion. Other studies have also showed that the pathological features in EOPC patients are similar to those seen in older PDAC patients, [8, 9] although the presence of more histological variants, especially mucinous carcinomas, has been observed in EOPC cases [8]. Interestingly, a lower rate of KRAS mutations in EOPC patients was found in two studies, [9, 43], although both were performed on a small number of cases (five and seven, respectively).
In our data, EOPC patients presented at a more advanced stage compared to older PDAC group (77% vs 56%, p = 0.015) but they were more frequently fit for adjuvant or palliative treatment (60% vs 46%, p = 0.008). While the overall survival of EOPC was similar to older PDAC patients with no statistically significant difference, EOPC patients who underwent surgical resection had a longer median overall survival of 25 months compared to 13 months for the same PDAC subpopulation. A similar finding has been observed in previous studies, with the highest survival of 41.8 months for resected cases reported by Duffy et al. [10] which has been attributed to young people having fewer comorbidities and being more suitable candidates for surgery and adjuvant chemotherapy [10, 12, 13, 36, 37]. Furthermore, He et al. also showed that EOPC patients had fewer post-operative complications [12] and McWilliams et al., [14] attributed the better survival rate among young people to a multitude of factors, including race, sex, year of diagnosis, stage of disease, tumour location and treatment [14].
Our study adds to a growing body of literature on the demographic and clinicopathological characteristics of EOPC patients, using contemporary classification and staging manuals. There are some limitations to our study: firstly, the disproportionate sample size of the two comparison groups, EOPC and older PDAC, although reflecting the general incidence of the disease and unavoidable, may have contributed to statistical errors in our analysis. In addition, the retrospective nature of the study has its own pitfalls, which include possible omission of patients that did not have a tissue diagnosis, and review of medical records with sometimes incomplete data. Finally, the study was conducted in a single tertiary expert centre with referred cases and may over-represent the patients that were suitable to undergo tissue biopsy prior to receiving more aggressive treatment. Enlarging the data set through a multi-centre collaborative approach might produce more robust results.

Conclusions

In conclusion, we present the data from a retrospective study of histologically confirmed PDAC patients over an 11-year period establishing for the first time the demographic and clinicopathological characteristics of EOPC in the multinational PDAC population inhabiting the greater London area. Our results showed demographic differences between EOPC and older PDAC patients, but no difference in association with any currently known risk factors for pancreatic cancer. EOPC patients who undergo surgery have a significantly better survival compared to their older counterparts, which reinforces the value of this therapeutic option, in combination with neoadjuvant chemotherapy/radiotherapy for downstaging of the disease. As risk factors reported previously and in our study do not point to any major differences in demographic and clinicopathological characteristics between EOPC and older PDAC patients, molecular investigations are warranted in order to understand the molecular bases for the occurrence of this highly aggressive malignancy in young populations.

Funding

The study was funded by Barts Charity. The funding body had no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.

Availability of data and materials

The datasets used and analysed during the current study are available from the corresponding author on reasonable request.
The study is based on the retrospective examination of the clinical notes and review of histology reports. No patient material was accessed or reviewed. Therefore, obtaining patient consent for use of tissue in such cases is not deemed necessary according to the Human Tissue Act. The study was approved by Queen Mary University of London Review Board and London-Brent Research Ethics Committee (Reference Number: 05/Q0408/65).
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
2.
Zurück zum Zitat Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.CrossRefPubMed Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–21.CrossRefPubMed
3.
Zurück zum Zitat Raimondi S, Maisonneuve P, Lowenfels AB. Epidemiology of pancreatic cancer: an overview. Nat Rev Gastroenterol Hepatol. 2009;6(12):699–708.CrossRefPubMed Raimondi S, Maisonneuve P, Lowenfels AB. Epidemiology of pancreatic cancer: an overview. Nat Rev Gastroenterol Hepatol. 2009;6(12):699–708.CrossRefPubMed
4.
Zurück zum Zitat Lin JC, Chan DC, Chen PJ, Chu HC, Chueh TH, Huang HH, et al. Clinical characteristics of early onset pancreatic adenocarcinoma: a medical center experience and review of the literature. Pancreas. 2011;40(4):638–9.CrossRefPubMed Lin JC, Chan DC, Chen PJ, Chu HC, Chueh TH, Huang HH, et al. Clinical characteristics of early onset pancreatic adenocarcinoma: a medical center experience and review of the literature. Pancreas. 2011;40(4):638–9.CrossRefPubMed
5.
Zurück zum Zitat Tingstedt B, Weitkamper C, Andersson R. Early onset pancreatic cancer: a controlled trial. Ann Gastroenterol. 2011;24(3):206–12.PubMedPubMedCentral Tingstedt B, Weitkamper C, Andersson R. Early onset pancreatic cancer: a controlled trial. Ann Gastroenterol. 2011;24(3):206–12.PubMedPubMedCentral
6.
Zurück zum Zitat Beeghly-Fadiel A, Luu HN, Du L, Shi C, McGavic DP, Parikh AA, et al. Early onset pancreatic malignancies: clinical characteristics and survival associations. Int J Cancer. 2016;139(10):2169–77.CrossRefPubMed Beeghly-Fadiel A, Luu HN, Du L, Shi C, McGavic DP, Parikh AA, et al. Early onset pancreatic malignancies: clinical characteristics and survival associations. Int J Cancer. 2016;139(10):2169–77.CrossRefPubMed
7.
Zurück zum Zitat Piciucchi M, Capurso G, Valente R, Larghi A, Archibugi L, Signoretti M, et al. Early onset pancreatic cancer: risk factors, presentation and outcome. Pancreatology. 2015;15(2):151–5.CrossRefPubMed Piciucchi M, Capurso G, Valente R, Larghi A, Archibugi L, Signoretti M, et al. Early onset pancreatic cancer: risk factors, presentation and outcome. Pancreatology. 2015;15(2):151–5.CrossRefPubMed
8.
Zurück zum Zitat Luttges J, Stigge C, Pacena M, Kloppel G. Rare ductal adenocarcinoma of the pancreas in patients younger than age 40 years. Cancer. 2004;100(1):173–82.CrossRefPubMed Luttges J, Stigge C, Pacena M, Kloppel G. Rare ductal adenocarcinoma of the pancreas in patients younger than age 40 years. Cancer. 2004;100(1):173–82.CrossRefPubMed
9.
Zurück zum Zitat Bergmann F, Aulmann S, Wente MN, Penzel R, Esposito I, Kleeff J, et al. Molecular characterisation of pancreatic ductal adenocarcinoma in patients under 40. J Clin Pathol. 2006;59(6):580–4.CrossRefPubMedPubMedCentral Bergmann F, Aulmann S, Wente MN, Penzel R, Esposito I, Kleeff J, et al. Molecular characterisation of pancreatic ductal adenocarcinoma in patients under 40. J Clin Pathol. 2006;59(6):580–4.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Duffy A, Capanu M, Allen P, Kurtz R, Olson SH, Ludwig E, et al. Pancreatic adenocarcinoma in a young patient population--12-year experience at memorial Sloan Kettering Cancer center. J Surg Oncol. 2009;100(1):8–12.CrossRefPubMed Duffy A, Capanu M, Allen P, Kurtz R, Olson SH, Ludwig E, et al. Pancreatic adenocarcinoma in a young patient population--12-year experience at memorial Sloan Kettering Cancer center. J Surg Oncol. 2009;100(1):8–12.CrossRefPubMed
11.
Zurück zum Zitat Liszka L, Pajak J, Mrowiec S, Zielinska-Pajak E, Golka D, Lampe P. Precursor lesions of early onset pancreatic cancer. Virchows Arch. 2011;458(4):439–51.CrossRefPubMedPubMedCentral Liszka L, Pajak J, Mrowiec S, Zielinska-Pajak E, Golka D, Lampe P. Precursor lesions of early onset pancreatic cancer. Virchows Arch. 2011;458(4):439–51.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat He J, Edil BH, Cameron JL, Schulick RD, Hruban RH, Herman JM, et al. Young patients undergoing resection of pancreatic cancer fare better than their older counterparts. J Gastrointest Surg. 2013;17(2):339–44.CrossRefPubMed He J, Edil BH, Cameron JL, Schulick RD, Hruban RH, Herman JM, et al. Young patients undergoing resection of pancreatic cancer fare better than their older counterparts. J Gastrointest Surg. 2013;17(2):339–44.CrossRefPubMed
13.
Zurück zum Zitat Jiang QL, Zhang SS, Chen YT, Zhang JW, Wang CF. Risk factors for early-onset pancreatic cancer patients, and survival analysis. Int J Clin Exp Med. 2017;10(6):9416–23. Jiang QL, Zhang SS, Chen YT, Zhang JW, Wang CF. Risk factors for early-onset pancreatic cancer patients, and survival analysis. Int J Clin Exp Med. 2017;10(6):9416–23.
14.
Zurück zum Zitat McWilliams RR, Maisonneuve P, Bamlet WR, Petersen GM, Li D, Risch HA, et al. Risk factors for early-onset and very-early-onset pancreatic adenocarcinoma: a pancreatic Cancer case-control consortium (PanC4) analysis. Pancreas. 2016;45(2):311–6.CrossRefPubMedPubMedCentral McWilliams RR, Maisonneuve P, Bamlet WR, Petersen GM, Li D, Risch HA, et al. Risk factors for early-onset and very-early-onset pancreatic adenocarcinoma: a pancreatic Cancer case-control consortium (PanC4) analysis. Pancreas. 2016;45(2):311–6.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Lynch HT, Smyrk T, Kern SE, Hruban RH, Lightdale CJ, Lemon SJ, et al. Familial pancreatic cancer: a review. Semin Oncol. 1996;23(2):251–75.PubMed Lynch HT, Smyrk T, Kern SE, Hruban RH, Lightdale CJ, Lemon SJ, et al. Familial pancreatic cancer: a review. Semin Oncol. 1996;23(2):251–75.PubMed
16.
Zurück zum Zitat Tersmette AC, Petersen GM, Offerhaus GJ, Falatko FC, Brune KA, Goggins M, et al. Increased risk of incident pancreatic cancer among first-degree relatives of patients with familial pancreatic cancer. Clin Cancer Res. 2001;7(3):738–44.PubMed Tersmette AC, Petersen GM, Offerhaus GJ, Falatko FC, Brune KA, Goggins M, et al. Increased risk of incident pancreatic cancer among first-degree relatives of patients with familial pancreatic cancer. Clin Cancer Res. 2001;7(3):738–44.PubMed
17.
Zurück zum Zitat James TA, Sheldon DG, Rajput A, Kuvshinoff BW, Javle MM, Nava HR, et al. Risk factors associated with earlier age of onset in familial pancreatic carcinoma. Cancer. 2004;101(12):2722–6.CrossRefPubMed James TA, Sheldon DG, Rajput A, Kuvshinoff BW, Javle MM, Nava HR, et al. Risk factors associated with earlier age of onset in familial pancreatic carcinoma. Cancer. 2004;101(12):2722–6.CrossRefPubMed
18.
Zurück zum Zitat Kharazmi E, Fallah M, Sundquist K, Hemminki K. Familial risk of early and late onset cancer: nationwide prospective cohort study. BMJ. 2012;345:e8076.CrossRefPubMedPubMedCentral Kharazmi E, Fallah M, Sundquist K, Hemminki K. Familial risk of early and late onset cancer: nationwide prospective cohort study. BMJ. 2012;345:e8076.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Lowenfels AB, Maisonneuve P, Cavallini G, Ammann RW, Lankisch PG, Andersen JR, et al. Pancreatitis and the risk of pancreatic cancer. International pancreatitis study group. N Engl J Med. 1993;328(20):1433–7.CrossRefPubMed Lowenfels AB, Maisonneuve P, Cavallini G, Ammann RW, Lankisch PG, Andersen JR, et al. Pancreatitis and the risk of pancreatic cancer. International pancreatitis study group. N Engl J Med. 1993;328(20):1433–7.CrossRefPubMed
20.
Zurück zum Zitat Lowenfels AB, Maisonneuve P, DiMagno EP, Elitsur Y, Gates LK Jr, Perrault J, et al. Hereditary pancreatitis and the risk of pancreatic cancer. International hereditary pancreatitis study group. J Natl Cancer Inst. 1997;89(6):442–6.CrossRefPubMed Lowenfels AB, Maisonneuve P, DiMagno EP, Elitsur Y, Gates LK Jr, Perrault J, et al. Hereditary pancreatitis and the risk of pancreatic cancer. International hereditary pancreatitis study group. J Natl Cancer Inst. 1997;89(6):442–6.CrossRefPubMed
21.
Zurück zum Zitat Brand RE, Lynch HT. Hereditary pancreatic adenocarcinoma. A clinical perspective. Med Clin North Am. 2000;84(3):665–75.CrossRefPubMed Brand RE, Lynch HT. Hereditary pancreatic adenocarcinoma. A clinical perspective. Med Clin North Am. 2000;84(3):665–75.CrossRefPubMed
22.
Zurück zum Zitat Kastrinos F, Mukherjee B, Tayob N, Wang F, Sparr J, Raymond VM, et al. Risk of pancreatic cancer in families with lynch syndrome. JAMA. 2009;302(16):1790–5.CrossRefPubMedPubMedCentral Kastrinos F, Mukherjee B, Tayob N, Wang F, Sparr J, Raymond VM, et al. Risk of pancreatic cancer in families with lynch syndrome. JAMA. 2009;302(16):1790–5.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Bowlby LS. Pancreatic adenocarcinoma in an adolescent male with Peutz-Jeghers syndrome. Hum Pathol. 1986;17(1):97–9.CrossRefPubMed Bowlby LS. Pancreatic adenocarcinoma in an adolescent male with Peutz-Jeghers syndrome. Hum Pathol. 1986;17(1):97–9.CrossRefPubMed
24.
Zurück zum Zitat Thatcher BS, May ES, Taxier MS, Bonta JA, Murthy L. Pancreatic adenocarcinoma in a patient with Peutz-Jeghers syndrome--a case report and literature review. Am J Gastroenterol. 1986;81(7):594–7.PubMed Thatcher BS, May ES, Taxier MS, Bonta JA, Murthy L. Pancreatic adenocarcinoma in a patient with Peutz-Jeghers syndrome--a case report and literature review. Am J Gastroenterol. 1986;81(7):594–7.PubMed
25.
Zurück zum Zitat Everhart J, Wright D. Diabetes mellitus as a risk factor for pancreatic cancer. A meta-analysis JAMA. 1995;273(20):1605–9.PubMed Everhart J, Wright D. Diabetes mellitus as a risk factor for pancreatic cancer. A meta-analysis JAMA. 1995;273(20):1605–9.PubMed
26.
Zurück zum Zitat Luo J, Iwasaki M, Inoue M, Sasazuki S, Otani T, Ye W, et al. Body mass index, physical activity and the risk of pancreatic cancer in relation to smoking status and history of diabetes: a large-scale population-based cohort study in Japan--the JPHC study. Cancer Causes Control. 2007;18(6):603–12.CrossRefPubMed Luo J, Iwasaki M, Inoue M, Sasazuki S, Otani T, Ye W, et al. Body mass index, physical activity and the risk of pancreatic cancer in relation to smoking status and history of diabetes: a large-scale population-based cohort study in Japan--the JPHC study. Cancer Causes Control. 2007;18(6):603–12.CrossRefPubMed
27.
Zurück zum Zitat Luo J, Nordenvall C, Nyren O, Adami HO, Permert J, Ye W. The risk of pancreatic cancer in patients with gastric or duodenal ulcer disease. Int J Cancer. 2007;120(2):368–72.CrossRefPubMed Luo J, Nordenvall C, Nyren O, Adami HO, Permert J, Ye W. The risk of pancreatic cancer in patients with gastric or duodenal ulcer disease. Int J Cancer. 2007;120(2):368–72.CrossRefPubMed
28.
Zurück zum Zitat Raimondi S, Maisonneuve P, Lohr JM, Lowenfels AB. Early onset pancreatic cancer: evidence of a major role for smoking and genetic factors. Cancer Epidemiol Biomark Prev. 2007;16(9):1894–7.CrossRef Raimondi S, Maisonneuve P, Lohr JM, Lowenfels AB. Early onset pancreatic cancer: evidence of a major role for smoking and genetic factors. Cancer Epidemiol Biomark Prev. 2007;16(9):1894–7.CrossRef
30.
Zurück zum Zitat Brand RE, Greer JB, Zolotarevsky E, Brand R, Du H, Simeone D, et al. Pancreatic cancer patients who smoke and drink are diagnosed at younger ages. Clin Gastroenterol Hepatol. 2009;7(9):1007–12.CrossRefPubMedPubMedCentral Brand RE, Greer JB, Zolotarevsky E, Brand R, Du H, Simeone D, et al. Pancreatic cancer patients who smoke and drink are diagnosed at younger ages. Clin Gastroenterol Hepatol. 2009;7(9):1007–12.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Genkinger JM, Spiegelman D, Anderson KE, Bergkvist L, Bernstein L, van den Brandt PA, et al. Alcohol intake and pancreatic cancer risk: a pooled analysis of fourteen cohort studies. Cancer Epidemiol Biomark Prev. 2009;18(3):765–76.CrossRef Genkinger JM, Spiegelman D, Anderson KE, Bergkvist L, Bernstein L, van den Brandt PA, et al. Alcohol intake and pancreatic cancer risk: a pooled analysis of fourteen cohort studies. Cancer Epidemiol Biomark Prev. 2009;18(3):765–76.CrossRef
32.
Zurück zum Zitat Lin G, Zeng Z, Wang X, Wu Z, Wang J, Wang C, et al. Cholecystectomy and risk of pancreatic cancer: a meta-analysis of observational studies. Cancer Causes Control. 2012;23(1):59–67.CrossRefPubMed Lin G, Zeng Z, Wang X, Wu Z, Wang J, Wang C, et al. Cholecystectomy and risk of pancreatic cancer: a meta-analysis of observational studies. Cancer Causes Control. 2012;23(1):59–67.CrossRefPubMed
33.
Zurück zum Zitat Becker AE, Hernandez YG, Frucht H, Lucas AL. Pancreatic ductal adenocarcinoma: risk factors, screening, and early detection. World J Gastroenterol. 2014;20(32):11182–98.CrossRefPubMedPubMedCentral Becker AE, Hernandez YG, Frucht H, Lucas AL. Pancreatic ductal adenocarcinoma: risk factors, screening, and early detection. World J Gastroenterol. 2014;20(32):11182–98.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Gomez-Rubio P, Zock JP, Rava M, Marquez M, Sharp L, Hidalgo M, et al. Reduced risk of pancreatic cancer associated with asthma and nasal allergies. Gut. 2017;66(2):314–22.CrossRefPubMed Gomez-Rubio P, Zock JP, Rava M, Marquez M, Sharp L, Hidalgo M, et al. Reduced risk of pancreatic cancer associated with asthma and nasal allergies. Gut. 2017;66(2):314–22.CrossRefPubMed
35.
Zurück zum Zitat Edge SB, Compton CC. The American joint committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 2010;17(6):1471–4.CrossRefPubMed Edge SB, Compton CC. The American joint committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 2010;17(6):1471–4.CrossRefPubMed
36.
Zurück zum Zitat Raissouni S, Rais G, Mrabti H, Raissouni F, Mouzount H, Aitelhaj M, et al. Pancreatic adenocarcinoma in young adults in a moroccan population. J Gastrointest Cancer. 2012;43(4):607–11.CrossRefPubMed Raissouni S, Rais G, Mrabti H, Raissouni F, Mouzount H, Aitelhaj M, et al. Pancreatic adenocarcinoma in young adults in a moroccan population. J Gastrointest Cancer. 2012;43(4):607–11.CrossRefPubMed
37.
Zurück zum Zitat Ohmoto A, Yachida S, Kubo E, Takai E, Suzuki M, Shimada K, et al. Clinicopathologic features and germline sequence variants in young patients (</=40 years old) with pancreatic ductal adenocarcinoma. Pancreas. 2016;45(7):1056–61.CrossRefPubMed Ohmoto A, Yachida S, Kubo E, Takai E, Suzuki M, Shimada K, et al. Clinicopathologic features and germline sequence variants in young patients (</=40 years old) with pancreatic ductal adenocarcinoma. Pancreas. 2016;45(7):1056–61.CrossRefPubMed
38.
Zurück zum Zitat Miller BA, Kolonel LN, Bernstein L, Young Jr. JL, Swanson GM, West D, et al. Racial/Ethnic Patterns of Cancer in the United States 1988–1992, National Cancer Institute. Bethesda, MD, 1996.; 1996 1996. Contract No.: No. 96–4104. Miller BA, Kolonel LN, Bernstein L, Young Jr. JL, Swanson GM, West D, et al. Racial/Ethnic Patterns of Cancer in the United States 1988–1992, National Cancer Institute. Bethesda, MD, 1996.; 1996 1996. Contract No.: No. 96–4104.
39.
Zurück zum Zitat Howe GR, Jain M, Burch JD, Miller AB. Cigarette smoking and cancer of the pancreas: evidence from a population-based case-control study in Toronto. Canada Int J Cancer. 1991;47(3):323–8.CrossRefPubMed Howe GR, Jain M, Burch JD, Miller AB. Cigarette smoking and cancer of the pancreas: evidence from a population-based case-control study in Toronto. Canada Int J Cancer. 1991;47(3):323–8.CrossRefPubMed
40.
Zurück zum Zitat Silverman DT, Dunn JA, Hoover RN, Schiffman M, Lillemoe KD, Schoenberg JB, et al. Cigarette smoking and pancreas cancer: a case-control study based on direct interviews. J Natl Cancer Inst. 1994;86(20):1510–6.CrossRefPubMed Silverman DT, Dunn JA, Hoover RN, Schiffman M, Lillemoe KD, Schoenberg JB, et al. Cigarette smoking and pancreas cancer: a case-control study based on direct interviews. J Natl Cancer Inst. 1994;86(20):1510–6.CrossRefPubMed
41.
Zurück zum Zitat Soliman AS, El-Ghawalby N, Ezzat F, Bondy ML, Soultan A, Abdel-Wahab M, et al. Unusually high rate of young-onset pancreatic cancer in the East Nile Delta region of Egypt. Int J Gastrointest Cancer. 2002;32(2–3):143–51.CrossRefPubMed Soliman AS, El-Ghawalby N, Ezzat F, Bondy ML, Soultan A, Abdel-Wahab M, et al. Unusually high rate of young-onset pancreatic cancer in the East Nile Delta region of Egypt. Int J Gastrointest Cancer. 2002;32(2–3):143–51.CrossRefPubMed
42.
Zurück zum Zitat Bartsch DK, Gress TM, Langer P. Familial pancreatic cancer--current knowledge. Nat Rev Gastroenterol Hepatol. 2012;9(8):445–53.CrossRefPubMed Bartsch DK, Gress TM, Langer P. Familial pancreatic cancer--current knowledge. Nat Rev Gastroenterol Hepatol. 2012;9(8):445–53.CrossRefPubMed
43.
Zurück zum Zitat Del Chiaro M, Menicagli M, Campani D, Funel N, Pollina LE, Decarli N, et al. Sporadic pancreatic ductal carcinoma in patient aged less than 40 years. Pancreas. 2004;29(4):336. Del Chiaro M, Menicagli M, Campani D, Funel N, Pollina LE, Decarli N, et al. Sporadic pancreatic ductal carcinoma in patient aged less than 40 years. Pancreas. 2004;29(4):336.
Metadaten
Titel
Demographic, clinical, and pathological features of early onset pancreatic cancer patients
verfasst von
Chara Ntala
Silvana Debernardi
Roger M. Feakins
Tatjana Crnogorac-Jurcevic
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Gastroenterology / Ausgabe 1/2018
Elektronische ISSN: 1471-230X
DOI
https://doi.org/10.1186/s12876-018-0866-z

Weitere Artikel der Ausgabe 1/2018

BMC Gastroenterology 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

NSCLC: Progressionsfreies Überleben unter Osimertinib fast versiebenfacht

06.06.2024 ASCO 2024 Kongressbericht

Erste Ergebnisse der Phase-III-Studie LAURA etablieren Osimertinib als neuen Therapiestandard für Menschen mit nicht-resezierbarem, EGFR-mutiertem, nicht-kleinzelligem Lungenkarzinom im Stadium III, die nach definitiver Radiochemotherapie progressionsfrei sind. Auf der ASCO-Tagung wurden diese beeindruckenden Ergebnisse besprochen.

Hodgkin Lymphom: BrECADD-Regime übertrifft die Erwartungen

05.06.2024 ASCO 2024 Kongressbericht

Das Kombinationsregime BrECADD mit Brentuximab vedotin ermöglichte in der Studie HD21 beim fortgeschrittenen klassischen Hodgkin-Lymphom eine unerwartet hohe progressionsfreie Überlebensrate von 94,3% nach vier Jahren. Gleichzeitig war das Regime besser tolerabel als der bisherige Standard eBEACOPP.

Antikörper-Drug-Konjugat verdoppelt PFS bei Multiplem Myelom

05.06.2024 ASCO 2024 Nachrichten

Zwei Phase-3-Studien deuten auf erhebliche Vorteile des Antikörper-Wirkstoff-Konjugats Belantamab-Mafodotin bei vorbehandelten Personen mit Multiplem Myelom: Im Vergleich mit einer Standard-Tripeltherapie wurde das progressionsfreie Überleben teilweise mehr als verdoppelt.

Neuer TKI gegen CML: Höhere Wirksamkeit, seltener Nebenwirkungen

05.06.2024 Chronische myeloische Leukämie Nachrichten

Der Tyrosinkinasehemmer (TKI) Asciminib ist älteren Vertretern dieser Gruppe bei CML offenbar überlegen: Personen mit frisch diagnostizierter CML entwickelten damit in einer Phase-3-Studie häufiger eine gute molekulare Response, aber seltener ernste Nebenwirkungen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.