1. Patient characteristics
A total of 195 PDAC patients from multiple medical centers in China were enrolled in this retrospective study. The vast majority of patients were recruited from West China Medical Center between January 2016 and December 2019, and the rest came from the Cancer Hospital of Fudan University and the People's Hospital of Sichuan Province. Demographics and clinicopathological data of the study population were listed in Table 1. The median age of all patients was approximately 60 years (range: 27–79 years), and males were moderately overrepresented compared with females (56.4% vs 43.6%). Family history of any malignancy in first-degree relatives was noted in 31 patients (15.9%). 109 resectable patients (55.9%) and 85 unresectable patients (43.6%) received curative surgery and palliative surgery or just biopsy, respectively. At any point during the disease, 60 patients (30.8%) eventually had signs of metastasis. Limited-stage patients accouted for 36.7% of overall patients and 123 patients with advanced disease (62.8%) were included in this study.
Table 1
Baseline characteristics of overall patients. Abbreviations: mut, mutant; wt, wild-type; CA, carbohydrate antigen; DDR, DNA damage repair; N, node; N, number; T, tumor. P value was calculated by x2 except t test for age.
Variable
|
Overall cohort, N = 195
|
DDR status
|
p value
|
mut
N = 30 (15.4%)
|
wt
N = 165 (84.6%)
|
Age at diagnosis
|
|
|
|
0.35
|
Median(range), years
|
59.3
|
59.6
|
59.2
|
|
Sex, n(%)
|
|
|
|
0.569
|
Male
|
110(56.4%)
|
15(50%)
|
95(57.6%)
|
|
Female
|
85(43.6%)
|
15(50%)
|
70(42.4%)
|
|
Family history of cancer, n (%)
|
31(15.9%)
|
7(23.3%)
|
24(14.5%)
|
0.347
|
Pancreatic cancer
|
5(2.6%)
|
1(3.3%)
|
4(2.4%)
|
|
Any cancer
|
26(13.3%)
|
6(20%)
|
20(12.1%)
|
|
Location of primary tumor, n (%)
|
|
|
|
0.253
|
Head/Uncinate
|
123(63.1%)
|
15(50%)
|
108(65.5%)
|
|
Body/Tail
|
47(24.1%)
|
10(33.3%)
|
37(22.4%)
|
|
NA
|
24(12.3%)
|
5(16.7%)
|
19(11.5%)
|
|
Surgery, n (%)
|
|
|
|
0.674
|
R0 (Negative margin)
|
109(55.9%)
|
16(53.3%)
|
93(56.4%)
|
|
R1 (Positive margin)
|
83(42.6%)
|
13(43.3%)
|
70(42.4%)
|
|
NA
|
3(1.5%)
|
1(3.3%)
|
2(1.2%)
|
|
Pathological T stage, n (%)
|
|
|
|
0.596
|
T1 and T2
|
68(34.9%)
|
9(30.0%)
|
59(35.8%)
|
|
T3 and T4
|
124(63.6%)
|
20(66.7%)
|
104(63.0%)
|
|
NA
|
3(1.5%)
|
1(3.3%)
|
2(1.2%)
|
|
Pathological N stage, n (%)
|
|
|
|
0.47
|
N0
|
85(43.6%)
|
15(50.0%)
|
70(42.4%)
|
|
N1/N2
|
107(54.9%)
|
14(46.7%)
|
93(56.4%)
|
|
NA
|
3(1.5%)
|
1(3.3%)
|
2(1.2%)
|
|
Metastasis, n (%)
|
|
|
|
0.632
|
M0
|
132(67.7%)
|
19(63.3%)
|
112(67.9%)
|
|
M1
|
60(30.8%)
|
10(33.3%)
|
50(30.3%)
|
|
NA
|
3(1.5%)
|
1(3.3%)
|
2(1.2%)
|
|
Stage, n (%)
|
|
|
|
0.950
|
Limited stage
|
72(36.7%)
|
11(36.7%)
|
61(37.0%)
|
|
Advanced stage
|
123(62.8%)
|
19(63.3%)
|
104(63.0%)
|
|
Perivascular invasion, n (%)
|
|
|
|
0.686
|
Present
|
40(20.5%)
|
6(20.0%)
|
34(20.6%)
|
|
Absent
|
152(77.9%)
|
23(76.7%)
|
129(78.2%)
|
|
NA
|
3(1.5%)
|
1(3.3%)
|
2(1.2%)
|
|
Perineural invasion, n (%)
|
|
|
|
0.68
|
Present
|
75(38.5%)
|
11(36.7%)
|
64(38.8%)
|
|
Absent
|
117(60.0%)
|
18(60.0%)
|
99(60.0%)
|
|
NA
|
3(1.5%)
|
1(3.3%)
|
2(1.2%)
|
|
CA 19–9, n (%)
|
|
|
|
0.055
|
Normal
|
44(22.6%)
|
7(23.3%)
|
37(22.4%)
|
|
Elevated
|
145(74.4%)
|
20(66.7%)
|
125(75.8%)
|
|
Unknown
|
6(3.1%)
|
3(10.0%)
|
3(1.8%)
|
|
Surgery, n (%)
|
|
|
|
0.857
|
Curative surgery
|
109(55.9%)
|
16(53.3%)
|
93(56.4%)
|
|
Unresectable cancer
|
85(43.6%)
|
14(46.7%)
|
71(43.0%)
|
|
NA
|
1(0.5%)
|
0(0.0%)
|
1(0.6%)
|
|
Adjuvant radiotherap or chemotherapy, n (%)
|
|
|
|
0.094
|
Yes
|
84(43.1%)
|
15(50%)
|
69(41.8%)
|
|
No
|
44(22.5%)
|
7(23.3%)
|
37(22.4%)
|
|
Unknown
|
67(34.4%)
|
8(26.7%)
|
59(35.8%)
|
|
In total, thirty patients (15.4%) were identified as mutant germline or somatic DDR gene in our study by NGS. The remaining 165 patients (84.6%) were therefore confirmed as DDR wild-type genotype and were matched by several clinical characteristics to the DDR mutated patients (Table 1). Generally, no significant difference in baseline characteristics of patients between the two groups. For example, there were equal numbers of male and female patients in DDR mutated groups, and male patients were slightly overrepresented in the wild-type group (50% DDR mut vs 57.6% DDR wt, p = 0.569). The percentage of family cancer history was similar in each group, also for the presence of pancreatic cancer (3.3% DDR mut vs 2.4% DDR wt, p = 0.347). Regarding the TNM staging, the composition of each stage of patients was similar between the two groups (T1-T2: 30.0% DDR mut vs 35.8% DDR wt; T3-T4 66.7% DDR mut vs 63.0% DDR wt; p = 0.596). More importantly, there was no difference between patiens in limited stage or advanced stage (p = 0.950). In conclusion, these results showed that no significant difference in other variables between the two groups, except for the DDR gene mutation status.
2. Mutation profiles of main driver genes
We performed NGS for 195 Chinese PDAC patients enrolled in our study, which has revealed a complex mutational landscape about genes known to be important in pancreatic cancer. 565 deleterious mutations were detected in all patients. The average mutations per cancer sample were 2.9 and 13 patients (6.6%) did not have any alterations in the genes of our panel. The whole mutation landscape of our cohorts is illustrated in Figure 1. Kirsten-ras protein (KRAS) was the most prevalent mutating gene, which occurred in 83.6% patients of our cohorts. Other frequent genomic alterations were listed as follows: tumor protein p53 (TP53) (62.05% in our cohorts vs 51% in TCGA), cyclin-dependent kinase inhibitor 2A (CDKN2A) (27.18% vs 11%), drosophila mothers against decapentaplegic homolog 4 (SMAD4) (17.44% vs 15%). Next, we investigated the influence of mutations in driver genes to the clinical outcomes of advanced patients. KRAS mutated patients with significantly lower overall survival (OS) than wild-type patients (Figure 2A). Interestingly, in this study, no significant correlation was found between the number of drive gene mutations and OS (Figure 2B), which differed from the report of other studies that more drive gene mutations may lead to shorter survival in PDAC patients [14-15]. As the fifth most common genetic alteration, a handful of genes related to DNA damage repair were identified in 15.38% of patients in our study. Among the genetic alterations, BRCA2 germline mutation was the most prevalent mutation of DDR deficiency. BRCA2, ATM, RAD50 and MLH1 genes were mutated in 9 (4.62%), 8 (4.10%), 3 (1.54%) and 2 (1.03%) of all patients, respectively. Other mutant DDR genes, such as BRCA1, MSH, RAD51, PMS2, PALB2, FANCA, FANCE, BLM, CHEK2, and FANCD2, were found in one patient (0.51%), respectively (Figure 2C).
3. Survival analyses based on DDR mutation status
The mutation profiles of all DDR gene mutations detected in our study has shown in Fig. 3A. There were several different alteration types among these mutations, including missense, nonsense, frameshift, intron mutation, and copy number variation (CNV)-loss. The detailed mutational information (mutation level, amino acid change, and corresponding functions) was listed in Table 2. A total of 36 mutations of DDR genes were identified in 30 patients, including 19 somatic mutations and 17 germline mutations (Fig. 3B). Six (3.07%) patients had more than one mutation. We recorded 12 germline and somatic deleterious BRCA1/2 mutations in 9 patients, 1 of which (0.5%) occurred in BRCA1, and 11 (4.7%) occurred in BRCA2. Among them, 2 patients had 2 sites of BRCA2 mutation simultaneously.
Table 2
Mutation details of DNA damage repair gene in 30 patients of our cohort.
Patient ID
|
Age
|
Sex
|
Mut Level
|
Amino acid change
|
Function
|
Patient 167
|
M
|
55
|
ATM germline
|
p.R1882*
|
Nonsense
|
Patient 185
|
M
|
61
|
ATM germline
|
p.C1899*
|
Nonsense
|
Patient 186
|
F
|
71
|
ATM germline
|
p.K468Efs*18
|
Frameshift
|
Patient 187
|
F
|
71
|
ATM germline
|
p.Q441Afs*45
|
Frameshift
|
Patient 195
|
M
|
55
|
ATM somatic
|
p.K2811Sfs*46
|
Frameshift
|
Patient 189
|
F
|
63
|
ATM somatic
|
R1898*
|
Nonsense
|
Patient 190
|
F
|
69
|
ATM somatic
|
p.G509*, p.L1651*
|
Nonsense
|
Patient 188
|
M
|
63
|
ATM somatic
|
c.2921 + 1G > A
|
Intron mutation
|
Patient 170
|
F
|
54
|
BLM germline
|
p.L258Efs*7
|
Frameshift
|
Patient 183
|
F
|
45
|
BRCA1 somatic
|
p.R1443*
|
Nonsense
|
Patient 173
|
F
|
67
|
BRCA2 somatic
|
p.T3033Nfs*11, p.K437Ifs*22
|
Frameshift
|
Patient 182
|
F
|
34
|
BRCA2 somatic
|
p.R3128*
|
Nonsense
|
Patient 180
|
M
|
63
|
BRCA2 somatic
|
D2723N
|
Missense
|
Patient 181
|
M
|
55
|
BRCA2 somatic
|
p.R2494*
|
Nonsense
|
Patient 182
|
F
|
34
|
BRCA2 germline
|
p.N2137Kfs*29
|
Frameshift
|
Patient 177
|
M
|
55
|
BRCA2 germline
|
c.3847_3848del
|
Indel
|
Patient 184
|
F
|
49
|
BRCA2 germline
|
Y1894*
|
Nonsense
|
Patient 192
|
F
|
73
|
BRCA2 germline
|
p.Q1073Rfs*4
|
Frameshift
|
Patient
113
|
M
|
34
|
BRCA2 germline
|
p.V1283Kfs*2
|
Frameshift
|
Patient 172
|
F
|
68
|
FANCA somatic
|
-
|
CNV-amplification
|
Patient 166
|
F
|
79
|
FANCE somatic
|
-
|
CNV-loss
|
Patient 191
|
M
|
60
|
FANCD2 germline
|
p.Q718*
|
Nonsense
|
Patient 174
|
M
|
47
|
MLH1 somatic
|
p.N287Kfs*10
|
Frameshift
|
Patient 175
|
M
|
57
|
MLH1 somatic
|
-
|
CNV-loss
|
Patient 194
|
M
|
62
|
MSH2 somatic
|
p.N566Ifs*24
|
Frameshift
|
Patient 194
|
M
|
62
|
MSH6 somatic
|
p.F1088Sfs*2
|
Frameshift
|
Patient 176
|
F
|
50
|
PALB2 somatic
|
p.F440Lfs*12
|
Frameshift
|
Patient 176
|
F
|
50
|
PALB2 germline
|
p.R753*
|
Nonsense
|
Patient 171
|
F
|
56
|
PMS2 somatic
|
-
|
CNV-loss
|
Patient 168
|
M
|
51
|
RAD50 germline
|
p.Q826*
|
Nonsense
|
Patient 169
|
M
|
67
|
RAD50 germline
|
c.3618 + 1G > A
|
Intron mutation
|
Patient 179
|
M
|
76
|
RAD50 germline
|
p.R1077*
|
Nonsense
|
Patient 171
|
F
|
56
|
RAD51 somatic
|
-
|
CNV-loss
|
Survival analyses were conducted to confirm the predictive and prognostic value of mutations in DDR-related genes. In our cohort, there were 123 patients (63.1%) in the advanced cohorts and 102 of advanced patients had survival data. Among them, 104 patients were DDR wildtype, while 19 patients were identified as DDR deficiency. The median OS of advanced patients was 11.69 months. The patients with DDR deficiency showed no benefit in OS compared to wild-type patients (p = 0.71) (Fig. 3C).
4. The effects of olaparib, platinum-based chemotherapy and PD-1/PD-L1 blockade on overall survival to patients with the DDR deficiency.
Of all the 195 patients, 22 have ever received any one of these DDR targeting drugs (olaparib, platinum-based chemotherapy and PD-1 blockades) and ten of them harbored DDR deficiency. Most patients who received these drugs harbored BRCA1/2 or ATM mutations (Fig. 4A). In the 18 advanced DDR-mutated patients, 4 patients received the second-line olaparib treatment after the failure of chemotherapy with gemicitabine and nab-paclitaxel or platinum. An improvement of OS was observed in the group with olaparib treatment compared to those without (p = 0.034; Fig. 4B).
There were overall 15 patients treated with platinum-based chemotherapy in our study, 9 of them were DDR wild-type while 6 were in the DDR mutated group. In advanced patients with DDR mutations, a total of 5 patients have received the platinum chemotherapy during the whole therapeutic course. 3 of them received second-line platinum-based chemotherapy, including 1 patient with mFOLFIRINOX (modified 5-Fluorouracil, leucovorin, irinotecan and oxaliplatin) regimen, after the tumor progression of gemicitabine plus nab-paclitaxel. The other two patients both received the gemcitabine and platinum chemotherapy as the first-line treatment. However, one patient had progression disease after 3 cycles of platinum chemotherapy and another patient had the disease recurrence in two years. In the advanced patients, platinum-based chemotherapy was also found to result in favorable OS (p = 0.0096, Fig. 4C). Next, we investigated the correlation between DDR deficiency and response to PD-1 inhibitors. Although PD-L1 overexpressed in tumors of 6 advanced patients, the efficacy of PD-1 blockades was a little disappointing: 1 patients with intact DDR genes had stable disease (SD), meanwhile, of the remaining 5 patients with DDR deficiency, 1 was evaluated as partial response (PR), 3 as SD, and 1 as progression disease (PD) (based on RECIST 1.1). However, in the advanced patients with DDR deficiency, the OS was not significantly prolonged after treatment of PD-1 blockades (p = 0.14; Fig. 4D).
13 advanced patients with DDR deficiency had the treatment and survival records. Detailed data of these individual patients were summarized in Fig. 5A. Matched therapy was defined as precise treatment according to the molecular profiling of the individual patient. For example, the matched therapy of DDR mutations included olaparib and platinum-based chemotherapy, and PD-1 blockade was matched therapy for positive PD-L1 expression. As shown in Fig. 5B, the participation of molecularly matched therapy in the treatment course significantly improved the overall survival of patients compared to those treated with unmatched therapy.
5. Correlations between hypermutation phenotype and DDR mutation
In our study, TMB could be evaluated in 87 patients who profiled by the “OK partner” panel. The median level of TMB was 4.9 mutations/Mb (range, 0.81–15.32 mutations/Mb). By analyzing the sequencing data of enrolled patients, we identified no significant difference of TMB between patients with DDR mutations and those in wild-type status (P = 0.384; Fig. 6A). However, in the DDR mutated group, a higher proportion of patients had medium or high level of TMB (56.25% DDR mut vs 38.23% DDR wt), and fewer patients were located at the low level of TMB (31.25% DDR mut vs 47.06% DDR wt).
In order to meet the need to response appropriately to different kinds of DNA damage, mammalian cells have evolved intricate DNA repair pathways to repair a large variety of structurally genotoxic damages: mismatch repair (MMR), base-excision repair (BER), nucleotide excision repair (NER), homologous recombination (HR), non-homologous DNA end joining (NHEJ) pathway, translesion synthesis (TLS), Fanconi anemia (FA) and checkpoint factors (CPF). In this study, the mutational genes were associated with five pathways (Fig. 3B). To further disclose the main contributing components affecting the connection between DDR mutations and TMB, we investigated whether the mutations among these pathways of DDR system may affect the TMB levels. As shown in Fig. 6B-E, patients with genetic alterations in CPF (p = 0.424), HRR (p = 0.590), and FA pathways (p = 0.099) failed to show significant differences with corresponding wide-type patients. However, NHEJ pathway alterations demonstrated a comparably higher level of TMB than the NHEJ wild-type groups (p < 0.001).
In our study, 89 of 195 patients had the available information of microsatellite status. One patient was evaluated as MSI-high by known NGS sequencing sites and another MSI-low was confirmed by immunohistochemistry (IHC) detection. And the remaining patients were all microsatellite stable (MSS). In contrast to our hypothesis, the two MSI patients were both DDR wild-type. IHC information of PD-L1 protein was available in 102 patients, and 23 of them (22.5%) were positive. In the DDR mutated group, the proportion of patients with PD-L1 overexpression was a little higher than that in the wild-type group (29.17% DDR mut vs 20.51% DDR wt) (Fig. 6F).