Abstract
The platinum-based chemotherapy regimen FOLFIRINOX (leucovorin, fluorouracil, irinotecan, and oxaliplatin) is currently used as a standard treatment for patients with unresectable pancreatic cancer. FOLFIRINOX is associated with severe toxicities, including neutropenia, febrile neutropenia, and anorexia; however, there are currently no reliable biomarkers to predict its efficacy and safety. Several studies of patients with various cancers have shown that tumor expression of excision repair cross-complementing (ERCC) proteins and glutathione S-transferase Pi (GSTPi) correlates with the response to platinum-based chemotherapies. Therefore, in this study, we examined the associations between expression of ERCC proteins and GSTPi and the safety and efficacy of FOLFIRINOX in 34 patients with unresectable pancreatic cancer. ERCC1, ERCC2, ERCC4, and GSTPi expression were examined by immunohistochemical staining of tumor specimens and the results were correlated with overall survival, progression-free survival, response rate, disease control rate, and the frequency of grade 3-4 neutropenia and non-hematologic toxicities. We found that ERCC1, ERCC2, ERCC4, and GSTPi were expressed in tumor samples from 64%, 24%, 18%, and 64% of patients, respectively. Notably, there were no statistically significant associations between the expression pattern of any of the proteins and either the clinical outcomes or the frequency of grade 3-4 neutropenia or grade 3-4 anorexia. Collectively, these data indicate that tumor expression of ERCC1, ERCC2, ERCC4, and GSTPi does not predict the safety or efficacy of FOLFIRINOX in patients with pancreatic cancer.
Keywords: Pancreatic cancer, FOLFIRINOX, predictive factor, ERCC1, ERCC2, ERCC4, GSTPi, immunohistochemistry, biomarker
Introduction
The effectiveness of the platinum-based regimen FOLFIRINOX (oxaliplatin, irinotecan, fluorouracil, and leucovorin) as first-line chemotherapy in patients with metastatic pancreatic cancer was first reported in 2011 [1]. In Japan, FOLFIRINOX obtained regulatory approval for the treatment of pancreatic cancer in December 2013. FOLFIRINOX and nab-paclitaxel plus gemcitabine are both currently employed as standard chemotherapy regimens for unresectable pancreatic cancer in patients with good Eastern Cooperative Oncology Group performance status (ECOG PS) [2].
FOLFIRINOX is associated with severe toxicities, including neutropenia, febrile neutropenia, thrombocytopenia, anorexia, fatigue, and diarrhea, which frequently require dose reduction or treatment interruption. A phase 2 study of FOLFIRINOX in Japanese patients with pancreatic cancer reported high frequencies of toxicities, including grade 3 or 4 neutropenia (77.8% of patients), febrile neutropenia (22.2%), and anorexia (11.1%) [3]. These side effects could lead limited use of FOLFIRINOX. Recently, a modified FOLFIRINOX regimen showed satisfactory efficacy with less toxicity in pancreatic cancer patients [4-6]. However, there have been no direct comparisons of the safety and efficacy of FOLFIRINOX, modified FOLFIRINOX, and nab-paclitaxel plus gemcitabine, making it difficult to select the optimal treatment regimen for pancreatic cancer patients. There is thus great interest in discovering biomarkers that can predict the efficacy and safety of FOLFIRINOX in this disease.
DNA repair is an important role in resistance to platinum agents that act inhibiting DNA replication and transcription by binding to a DNA molecule in the form of platinum-DNA-adducts [7]. Excision repair cross-complementing (ERCC) protein 1, ERCC2, and ERCC4 are key proteins involved in the nucleotide excision repair pathway, which is a major component of the DNA repair response [8,9]. Previous work identified a correlation between expression of ERCC1 protein or mRNA, as measured by immunohistochemistry (IHC) or reverse-transcription polymerase chain reaction (RT-PCR) respectively, and the response to platinum agents, including oxaliplatin, in patients with gastrointestinal cancer, ovarian cancer, and non-small cell lung cancer (NSCLC) [10-19]. A single nucleotide polymorphism in ERCC2 (Lys751Gln) is associated with the survival of patients with colon cancer treated by oxaliplatin-based regimens [20], supporting the possible utility of ERCC proteins as biomarkers of the response to platinum chemotherapies. Another potential biomarker is glutathione S-transferase Pi (GSTPi, also known as GSTP1). GSTs are a family of detoxification enzymes found in all aerobic organisms and play protective roles in preventing cellular damage from toxic compounds, including chemotherapeutic agents [21]. Polymorphisms have been detected in many of the genes encoding the main GST isoenzymes, including GSTPi. One polymorphism in GSTPi is reportedly related to the sensitivity of colorectal cancer to platinum-based compounds [22].
There have been few reports examining the correlation between ERCC1, ERCC2, ERCC4, and GSTPi expression and the response to platinum-based chemotherapies in patients with pancreatic cancer [16,18,23]. To address this knowledge gap, we examined the expression of these four candidate biomarkers in pancreatic cancer specimens from patients treated with FOLFIRINOX and investigated their relationship to various clinicopathological factors, including patient outcomes.
Material and methods
Patients and study design
This was a single-center, retrospective cohort study of 34 patients with unresectable pancreatic cancer who started treatment with FOLFIRINOX between December 2013 and September 2015. All patients were selected for this study based on histologically confirmed pancreatic adenocarcinoma and the availability of adequate tissue derived from (i) endoscopic ultrasonography-guided fine-needle aspiration (EUS-FNA) of the primary pancreatic tumor or lymph node metastasis, (ii) fine-needle biopsy from liver metastasis, (iii) forceps biopsy of bile duct-invasive tumor or duodenum-invasive tumor, or (iv) previously resected surgical specimens from primary tumors. Clinical data were obtained by retrospective chart review. All patients had an ECOG PS score of 0 or 1 [2] and had adequate bone marrow and renal function. Evaluation of the response to chemotherapy (complete response [CR], partial response [PR], stable disease [SD], and progressive disease [PD]) was based on the Response Evaluation Criteria In Solid Tumors guidelines [24]. All patients provided informed consent. This study was approved by the Institutional Review Board of Kanagawa Cancer Center.
Immunohistochemistry
IHC was performed using standard procedures. In brief, formalin-fixed, paraffin-embedded tissue samples were cut into 4-μm-thick sections, deparaffinized and rehydrated. Endogenous peroxidase was blocked by incubation with 3% hydrogen peroxide for 10 min at room temperature, and antigen retrieval was achieved by incubating sections for 15 min at 121°C in citrate buffer (pH 6.0). Sections were then incubated with one of the following primary antibodies for 60 min at room temperature: anti-ERCC1 (#12345, Cell Signaling Technology, Danvers, MA, USA, 1:100 dilution), anti-ERCC2 (10818-1-AP, Proteintech, Rosemont, IL, USA, 1:75), anti-ERCC4 (IHC-00263, Bethyl Laboratories, Montgomery, TX, USA, 1:300), and anti-GSTP1 (#3369, Cell Signaling Technology, 1:800). After washing, sections were incubated for 30 min at room temperature with secondary antibody and polymer detection system (N-Histofine, Nichirei Biosciences, Tokyo, Japan) and then stained with chromogen (DAB Chromogen Kit, Nichirei Biosciences). Finally, the sections were counterstained with hematoxylin and dehydrated for mounting.
Evaluation of immunohistochemical staining
Protein staining was assessed independently by two investigators who were blinded to the patients’ identities and clinical outcomes. If the investigators’ opinions differed, the final result was decided by consensus. Slides were analyzed by light microscopy. Staining was scored for intensity (1, weak; 2, moderate; 3, strong) and the percentage of cells positively stained (1, 1% to 9.9%; 2, 10% to <49.9%; 3, 50% to 100%). ERCC1, ERCC2, and ERCC4 expression was considered to be positive when the intensity and percentage scores were 2 or 3 and the tumor cells showed nuclear staining. GSTPi expression was considered to be positive when the intensity and percentage scores were 2 or 3 and the tumor cells showed nuclear or cytoplasmic staining. All other staining patterns were considered negative. This evaluation system was reported in the past study [10].
Clinical endpoints and statistical analysis
The clinical outcomes investigated were overall survival (OS), progression-free survival (PFS), response rate (RR), disease control rate (DCR), and the frequency of grade 3-4 neutropenia and non-hematologic toxicities. Associations between protein expression and OS or PFS were analyzed using the Kaplan-Meier method and evaluated using the log-rank test. Relationships between protein expression and RR, DCR, grade 3-4 neutropenia frequency, and grade 3-4 anorexia frequency were evaluated by Fisher’s exact test. Multivariate Cox regression analysis was used to evaluate relationships between clinical outcomes and protein expression or clinicopathological variables, such as ECOG PS [2] and neutrophil-to-lymphocyte ratio [25], which are known predictive factors in several malignancies.
Continuous variables were expressed as the mean and 95% confidence interval (CI) for multivariate Cox regression analysis or the median and range. Categorical variables were expressed as counts and percentages, and differences in categorical variables were analyzed using Fisher’s exact test. A P value <0.05 was regarded as statistically significant. Statistical analyses were performed using JMP Pro version 12.2.0 (SAS Institute Inc., Cary, NC, USA).
Results
Patient characteristics
We enrolled 56 patients with unresectable pancreatic cancer who started treatment with FOLFIRINOX at our institution between December 2013 and September 2015. Thirty-four of the 56 patients had adequate tissue specimens available for IHC and were included in the study. The median age of the 34 patients was 61 years (range, 42-73 years). The patient characteristics are presented in Table 1.
Table 1.
Clinical characteristics of the patients (n = 34)
| N (%) | |
|---|---|
| Age, median (range) | 61 (42-73) |
| Gender | |
| Male | 20 (59) |
| Female | 14 (41) |
| Line | |
| 1st line therapy | 26 (76) |
| 2nd line therapy | 3 (9) |
| 3rd line therapy | 5 (15) |
| UICC Stage | |
| III | 3 (9) |
| IV | 31 (91) |
| Performance status (ECOG) | |
| 0 | 16 (47) |
| 1 | 18 (53) |
| Initial dose modification | 17 (50) |
| Neutrophil-to-lymphocyte ratio ≥5 | 8 (24) |
Abbreviations: ECOG, Eastern Cooperative Oncology Group; UICC, Union for International Cancer Control.
Expression of biologic markers and relationships to clinical outcomes
Of the 34 specimens collected, 13 were from primary pancreatic tumors, 11 were from liver metastases, 3 were from duodenum-invasive tumors, 2 were from bile duct-invasive tumors, and 1 was from a lymph node metastasis. The four remaining samples were obtained from previously resected surgical specimens.
IHC analysis revealed that ERCC1 was expressed in 22 of the 34 specimens (64%) examined and ERCC2, ERCC4, and GSTPi were expressed in 24%, 18%, and 64% of specimens, respectively (Figure 1). ERCC1 and ERCC4, which form an enzyme complex in the nucleotide excision repair pathway, were co-expressed in 6 samples (18%). ERCC1, ERCC2, ERCC4, and GSTPi were all co-expressed in only 2 of the 34 samples (6%). None of the proteins were detected by IHC in normal (non-tumor) cells.
Figure 1.

Immunohistochemical staining of patient specimens. A-D. Representative images of immunohistochemical staining of the four proteins to illustrate typical intensity and percentage staining scores (both on a scale of 1-3). A. Excision repair cross-complementing (ERCC): intensity score 1, percentage score 3. B. ERCC2: intensity score 3, percentage score 2. C. ERCC4: intensity score 3, percentage score 2. D. Glutathione S-transferase Pi, intensity score 3, percentage score 3. Scale bar = 500 μm.
The patient clinical outcomes are summarized in Table 2. There were no significant differences between outcomes in patients with positive or negative expression of ERCC1, ERCC2, ERCC4, or GSTPi. Kaplan-Meier OS and PFS curves stratified according to protein expression are shown in Figure 2. Although the differences in OS and PFS between patients with positive vs. negative protein expression were not statistically significant, the median PFS tended to be longer for patients expressing ERCC1 (154.5 days vs. 92 days, P = 0.266; Figure 2A) and ERCC2 (252 days vs. 92 days, P = 0.222; Figure 2B). Similarly, we observed no significant differences in RR or DCR between patients with positive or negative expression of any of the proteins, but RR tended to be better for patients expressing ERCC1 (23% vs. 0%, P = 0.095; Table 2). Finally, we did not detect any significant correlations between the protein expression pattern and the frequency of either grade 3-4 neutropenia or grade 3-4 anorexia (Table 3).
Table 2.
Relationship between expression of biological markers and clinical outcomes
| N (%) | OS [95% CI] (days) | PFS [95% CI] (days) | RR (%) | DCR (%) | ||
|---|---|---|---|---|---|---|
| ERCC1 | Positive | 22 (64) | 391 [177, 616] | 154.5 [57, 270] | 23 | 64 |
| Negative | 12 (36) | 203.5 [123, 561] | 92 [30, 160] | 0 | 67 | |
| P-value | 0.503 | 0.266 | 0.095 | 0.583 | ||
| ERCC2 | Positive | 8 (24) | 399.5 [123, 821] | 252 [47, 342] | 13 | 75 |
| Negative | 26 (76) | 217.5 [162, 561] | 92 [57, 160] | 15 | 62 | |
| P-value | 0.623 | 0.222 | 0.666 | 0.402 | ||
| ERCC4 | Positive | 6 (18) | 216 [124, 821] | 178.5 [35, 309] | 17 | 67 |
| Negative | 28 (82) | 391 [177, 497] | 127 [57, 182] | 14 | 64 | |
| P-value | 0.750 | 0.911 | 0.647 | 0.649 | ||
| GSTPi | Positive | 22 (64) | 240 [162, 489] | 97 [48, 270] | 9 | 64 |
| Negative | 12 (36) | 391 [124, 616] | 129 [36, 182] | 25 | 67 | |
| P-value | 0.882 | 0.362 | 0.225 | 0.583 |
Abbreviations: CI, confidence interval; DCR, disease control rate; ERCC, excision repair cross-complementing; GSTPi, glutathione S-transferase Pi; OS, overall survival; PFS, progression-free survival; RR, response rate.
Figure 2.
Kaplan-Meier curves for patient survival. (A-D) Overall survival (OS) and progression-free survival (PFS) of patients stratified by tumor expression of excision repair cross-complementing (ERCC) 1 (A), ERCC2 (B), ERCC4 (C), and glutathione S-transferase Pi (GSTPi) (D).
Table 3.
Associations between the expression of biological markers and the frequency of grade 3-4 neutropenia and anorexia
| Grade 3-4 neutropenia (%) | Grade 3-4 anorexia (%) | ||
|---|---|---|---|
| ERCC1 | Positive | 91 | 14 |
| Negative | 75 | 8 | |
| P-value | 0.225 | 0.556 | |
| ERCC2 | Positive | 88 | 13 |
| Negative | 85 | 12 | |
| P-value | 0.666 | 0.678 | |
| ERCC4 | Positive | 100 | 0 |
| Negative | 82 | 14 | |
| P-value | 0.353 | 0.441 | |
| GSTPi | Positive | 86 | 14 |
| Negative | 83 | 8 | |
| P-value | 0.590 | 0.556 |
Abbreviations: ERCC, excision repair cross-complementing; GSTPi, glutathione S-transferase Pi.
To further assess the possible associations of clinicopathological factors on OS and PFS, we performed multivariate Cox regression analysis (Tables 4 and 5, respectively). These analyses revealed that an ECOG PS of 1 was significantly associated with OS (hazard ratio [HR]: 9.50, 95% CI: 2.25-40.07, P = 0.002) and PFS (HR: 4.95, 95% CI: 1.75-14.00, P = 0.003). Similarly, a neutrophil-to-lymphocyte ratio ≥5 was significantly related to both OS (HR: 9.50, 95% CI: 2.25-40.07, P = 0.002 and HR: 6.02, 95% CI: 1.20-30.29, P = 0.03).
Table 4.
Multivariate Cox regression analysis of factors associated with overall survival
| Hazard ratio | 95% CI | P-value | ||
|---|---|---|---|---|
| ERCC1 | Positive | 0.444 | 0.112-1.762 | 0.248 |
| Negative | 1 | |||
| ERCC2 | Positive | 1.102 | 0.258-4.711 | 0.895 |
| Negative | 1 | |||
| ERCC4 | Positive | 3.294 | 0.653-17.103 | 0.156 |
| Negative | 1 | |||
| GSTPi | Positive | 2.899 | 0.458-18.357 | 0.258 |
| Negative | 1 | |||
| 2nd or 3rd line therapy | 2.171 | 0.370-12.726 | 0.390 | |
| 1st line therapy | 1 | |||
| UICC Stage | IV | 1.049 | 0.080-13.803 | 0.971 |
| III | 1 | |||
| Performance status (ECOG) | 1 | 9.502 | 2.254-40.067 | 0.002 |
| 0 | 1 | |||
| Neutrophil-to-lymphocyte ratio | ≥5 | 6.016 | 1.195-30.292 | 0.030 |
| <5 | 1 | |||
| Initial dose modification | 1.509 | 0.300-7.586 | 0.618 | |
| Original dose | 1 |
Abbreviations: CI, confidence interval; ECOG, Eastern Cooperative Oncology Group; ERCC, excision repair cross-complementing; GSTPi, glutathione S-transferase Pi; UICC, Union for International Cancer Control.
Table 5.
Multivariate Cox regression analysis of factors associated with progression-free survival
| Hazard ratio | 95% CI | P-value | ||
|---|---|---|---|---|
| ERCC1 | Positive | 0.371 | 0.123-1.119 | 0.078 |
| Negative | 1 | |||
| ERCC2 | Positive | 0.971 | 0.294-3.205 | 0.962 |
| Negative | 1 | |||
| ERCC4 | Positive | 2.150 | 0.539-8.579 | 0.278 |
| Negative | 1 | |||
| GSTPi | Positive | 1.946 | 0.608-6.228 | 0.262 |
| Negative | 1 | |||
| 2nd or 3rd line therapy | 2.556 | 0.782-8.356 | 0.120 | |
| 1st line therapy | 1 | |||
| UICC Stage | IV | 0.693 | 0.130-3.702 | 0.668 |
| III | 1 | |||
| Performance status (ECOG) | 1 | 4.947 | 1.748-14.000 | 0.003 |
| 0 | 1 | |||
| Neutrophil-to-lymphocyte ratio | ≥5 | 1.653 | 0.552-5.230 | 0.393 |
| <5 | 1 | |||
| Initial dose modification | 3.373 | 0.918-12.391 | 0.067 | |
| Original dose | 1 | |||
Abbreviations: CI, confidence interval; ECOG, Eastern Cooperative Oncology Group; ERCC, excision repair cross-complementing; GSTPi, glutathione S-transferase Pi; UICC, Union for International Cancer Control.
Discussion
To our knowledge, this is the first study to examine the relationship between tumor expression of ERCC1, ERCC2, ERCC4, and GSTPi, as assessed by IHC, and the response to FOLFIRINOX in pancreatic cancer patients. We found that none of these proteins could serve as predictive biomarkers in our patient cohort. Although the results did not reach the level of statistical significance, however, we did observe a trend between positive tumor expression of ERCC1 and PFS and RR, and between expression of ERCC2 and PFS.
The use of FOLFIRINOX for pancreatic cancer has been limited by its severe toxicities combined with better patient responses to gemcitabine [1,3]. However, there have been no randomized controlled trials to directly compare the efficacy of FOLFIRINOX and nab-paclitaxel plus gemcitabine, both of which are standard chemotherapy regimens for unresectable pancreatic cancer patients with good ECOG PS. Moreover, there are no established predictive biomarkers for the response to FOLFIRINOX, with the exception of gender and serum CA19-9, which are positive predictors of the response [26]. There is thus an unmet need to identify reliable biomarkers that can predict patient response to FOLFIRINOX.
Several reports have identified a relationship between tumor ERCC1 expression detected by IHC and the response to platinum agents, including oxaliplatin, in patients with gastrointestinal cancer, ovarian cancer, and NSCLC [10,12,18,19]. In addition, ERCC1 mRNA expression levels have been reported to correlate with resistance to platinum agents in patients with esophageal, gastric, colon, and ovarian cancers and NSCLC [11,13-15,17,19]. Only one study in pancreatic cancer has demonstrated a significant relationship between ERCC1 expression and the response to FOLFIRINOX, and that involved measurement of ERCC1 mRNA, not protein, levels [16].
The results of the present study contrast with earlier data showing a correlation between high ERCC1 expression and resistance to platinum agents in gastrointestinal cancer, ovarian cancer, and NSCLC [10-19]. However, our findings are consistent with other studies reporting no relationship between ERCC1 expression and response to platinum agents in pancreatic cancer, colon cancer, and neuroendocrine tumors [27-30]. We speculate that these differences may be explained partly by the different types of cancer studied and partly by differences in methodology. The two previous studies using IHC to detect ERCC1 expression in pancreatic cancer concluded that ERCC1 levels did not correlate with the response to platinum agents [18,23]. In contrast, the single study examining ERCC1 mRNA expression in pancreatic cancer did find an association with the response to platinum agents [16]. Consistent with the earlier studies, we found no evidence of a significant relationship between ERCC1 protein expression, as detected by IHC, and the response of pancreatic cancer patients to FOLFIRINOX.
Our failure to detect a relationship between protein expression and FOLFIRINOX response could be due to the small number of tumor cells evaluated in the sections. Only four surgically resected specimens were available in this study, and the remaining 30 specimens were collected by EUS-FNA or biopsy of liver metastases, bile duct-invasive tumors, or duodenum-invasive tumors. Biopsied specimens, especially those obtained by EUS-FNA, are generally small, and pancreatic cancers have abundant fibrotic stroma [31]. A study of surgically resected pancreatic cancer specimens reported a significant correlation between high ERCC1 expression, as detected by IHC, and shorter OS and recurrence-free survival [32].
This study has some limitations. First, as mentioned above, the sample size was relatively small and may have been insufficient to detect positive associations. Second, we used only IHC to detect ERCC1, ERCC2, ERCC4, and GSTPi expression, and use of a complementary method, such as mRNA analysis by RT-PCR, may provide more robust data. Third, the proportion of tumors positive for ERCC2 (24%) and ERCC4 (18%) was low, which may have complicated the interpretation of results. Finally, the specificities of the antibodies used for IHC are unclear, potentially resulting in incorrect expression data [33,34].
Conclusions
In conclusion, this is the first report to demonstrate that tumor expression of ERCC1, ERCC2, ERCC4, and GSTPi in pancreatic cancer, as measured by IHC, is not significantly associated with the outcome of FOLFIRINOX treatment and cannot be used as predictive biomarkers in this setting. Further studies with larger sample sizes that include examination of both protein and mRNA levels will be necessary to clarify the predictive value of these proteins.
Acknowledgements
We thank Anne M. O’Rourke, PhD, from Edanz Group (www.edanzediting.com/ac) for editing a draft of this manuscript. Supported by Kanagawa Cancer Center-Kanagawa Cancer Research Institute joint research grant: 27-06.
Disclosure of conflict of interest
None.
References
- 1.Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardiere C, Bennouna J, Bachet JB, Khemissa-Akouz F, Pere-Verge D, Delbaldo C, Assenat E, Chauffert B, Michel P, Montoto-Grillot C, Ducreux M Groupe Tumeurs Digestives of Unicancer; PRODIGE Intergroup. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–1825. doi: 10.1056/NEJMoa1011923. [DOI] [PubMed] [Google Scholar]
- 2.Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET, Carbone PP. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 1982;5:649–655. [PubMed] [Google Scholar]
- 3.Okusaka T, Ikeda M, Fukutomi A, Ioka T, Furuse J, Ohkawa S, Isayama H, Boku N. Phase II study of FOLFIRINOX for chemotherapy-naive Japanese patients with metastatic pancreatic cancer. Cancer Sci. 2014;105:1321–1326. doi: 10.1111/cas.12501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Stein SM, James ES, Deng Y, Cong X, Kortmansky JS, Li J, Staugaard C, Indukala D, Boustani AM, Patel V, Cha CH, Salem RR, Chang B, Hochster HS, Lacy J. Final analysis of a phase II study of modified FOLFIRINOX in locally advanced and metastatic pancreatic cancer. Br J Cancer. 2016;114:737–743. doi: 10.1038/bjc.2016.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Li X, Ma T, Zhang Q, Chen YG, Guo CX, Shen YN, Sun PW, Li GG, Gao SL, Que RS, Lou JY, Yu RS, Yuan Y, Wei QC, Wei SM, Zhang Y, Zheng L, Bai XL, Liang TB. Modified-FOLFIRINOX in metastatic pancreatic cancer: a prospective study in Chinese population. Cancer Lett. 2017;406:22–26. doi: 10.1016/j.canlet.2017.07.012. [DOI] [PubMed] [Google Scholar]
- 6.Ozaka M, Ishii H, Sato T, Ueno M, Ikeda M, Uesugi K, Sata N, Miyashita K, Mizuno N, Tsuji K, Okusaka T, Furuse J. A phase II study of modified FOLFIRINOX for chemotherapy-naive patients with metastatic pancreatic cancer. Cancer Chemother Pharmacol. 2018;81:1017–1023. doi: 10.1007/s00280-018-3577-9. [DOI] [PubMed] [Google Scholar]
- 7.Altaha R, Liang X, Yu JJ, Reed E. Excision repair cross complementing-group 1: gene expression and platinum resistance. Int J Mol Med. 2004;14:959–970. [PubMed] [Google Scholar]
- 8.Fuss JO, Cooper PK. DNA repair: dynamic defenders against cancer and aging. PLoS Biol. 2006;4:e203. doi: 10.1371/journal.pbio.0040203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gillet LC, Scharer OD. Molecular mechanisms of mammalian global genome nucleotide excision repair. Chem Rev. 2006;106:253–276. doi: 10.1021/cr040483f. [DOI] [PubMed] [Google Scholar]
- 10.Kim SH, Kwon HC, Oh SY, Lee DM, Lee S, Lee JH, Roh MS, Kim DC, Park KJ, Choi HJ, Kim HJ. Prognostic value of ERCC1, thymidylate synthase, and glutathione S-transferase pi for 5-FU/oxaliplatin chemotherapy in advanced colorectal cancer. Am J Clin Oncol. 2009;32:38–43. doi: 10.1097/COC.0b013e31817be58e. [DOI] [PubMed] [Google Scholar]
- 11.Shirota Y, Stoehlmacher J, Brabender J, Xiong YP, Uetake H, Danenberg KD, Groshen S, Tsao-Wei DD, Danenberg PV, Lenz HJ. ERCC1 and thymidylate synthase mRNA levels predict survival for colorectal cancer patients receiving combination oxaliplatin and fluorouracil chemotherapy. J. Clin. Oncol. 2001;19:4298–4304. doi: 10.1200/JCO.2001.19.23.4298. [DOI] [PubMed] [Google Scholar]
- 12.Olaussen KA, Dunant A, Fouret P, Brambilla E, Andre F, Haddad V, Taranchon E, Filipits M, Pirker R, Popper HH, Stahel R, Sabatier L, Pignon JP, Tursz T, Le Chevalier T, Soria JC IALT Bio Investigators. DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N Engl J Med. 2006;355:983–991. doi: 10.1056/NEJMoa060570. [DOI] [PubMed] [Google Scholar]
- 13.Joshi MB, Shirota Y, Danenberg KD, Conlon DH, Salonga DS, Herndon JE 2nd, Danenberg PV, Harpole DH Jr. High gene expression of TS1, GSTP1, and ERCC1 are risk factors for survival in patients treated with trimodality therapy for esophageal cancer. Clin Cancer Res. 2005;11:2215–2221. doi: 10.1158/1078-0432.CCR-04-1387. [DOI] [PubMed] [Google Scholar]
- 14.Metzger R, Leichman CG, Danenberg KD, Danenberg PV, Lenz HJ, Hayashi K, Groshen S, Salonga D, Cohen H, Laine L, Crookes P, Silberman H, Baranda J, Konda B, Leichman L. ERCC1 mRNA levels complement thymidylate synthase mRNA levels in predicting response and survival for gastric cancer patients receiving combination cisplatin and fluorouracil chemotherapy. J. Clin. Oncol. 1998;16:309–316. doi: 10.1200/JCO.1998.16.1.309. [DOI] [PubMed] [Google Scholar]
- 15.Dabholkar M, Bostick-Bruton F, Weber C, Bohr VA, Egwuagu C, Reed E. ERCC1 and ERCC2 expression in malignant tissues from ovarian cancer patients. J Natl Cancer Inst. 1992;84:1512–1517. doi: 10.1093/jnci/84.19.1512. [DOI] [PubMed] [Google Scholar]
- 16.Strippoli A, Rossi S, Martini M, Basso M, D’Argento E, Schinzari G, Barile R, Cassano A, Barone C. ERCC1 expression affects outcome in metastatic pancreatic carcinoma treated with FOLFIRINOX: a single institution analysis. Oncotarget. 2016;7:35159–35168. doi: 10.18632/oncotarget.9063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Deng Q, Yang H, Lin Y, Qiu Y, Gu X, He P, Zhao M, Wang H, Xu Y, Lin Y, Jiang J, He J, Zhou JX. Prognostic value of ERCC1 mRNA expression in non-small cell lung cancer, breast cancer, and gastric cancer in patients from southern China. Int J Clin Exp Pathol. 2014;7:8312–8321. [PMC free article] [PubMed] [Google Scholar]
- 18.Ulker M, Duman BB, Sahin B, Gumurdulu D. ERCC1 and RRM1 as a predictive parameter for non-small cell lung, ovarian or pancreas cancer treated with cisplatin and/or gemcitabine. Contemp Oncol (Pozn) 2015;19:207–213. doi: 10.5114/wo.2015.52656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chen S, Zhang J, Wang R, Luo X, Chen H. The platinum-based treatments for advanced non-small cell lung cancer, is low/negative ERCC1 expression better than high/positive ERCC1 expression? A meta-analysis. Lung Cancer. 2010;70:63–70. doi: 10.1016/j.lungcan.2010.05.010. [DOI] [PubMed] [Google Scholar]
- 20.Park DJ, Stoehlmacher J, Zhang W, Tsao-Wei DD, Groshen S, Lenz HJ. A Xeroderma pigmentosum group D gene polymorphism predicts clinical outcome to platinum-based chemotherapy in patients with advanced colorectal cancer. Cancer Res. 2001;61:8654–8658. [PubMed] [Google Scholar]
- 21.Prade L, Huber R, Manoharan TH, Fahl WE, Reuter W. Structures of class pi glutathione S-transferase from human placenta in complex with substrate, transition-state analogue and inhibitor. Structure. 1997;5:1287–1295. doi: 10.1016/s0969-2126(97)00281-5. [DOI] [PubMed] [Google Scholar]
- 22.Stoehlmacher J, Park DJ, Zhang W, Groshen S, Tsao-Wei DD, Yu MC, Lenz HJ. Association between glutathione S-transferase P1, T1, and M1 genetic polymorphism and survival of patients with metastatic colorectal cancer. J Natl Cancer Inst. 2002;94:936–942. doi: 10.1093/jnci/94.12.936. [DOI] [PubMed] [Google Scholar]
- 23.Peters GJ, Avan A, Ruiz MG, Orsini V, Avan A, Giovannetti E, Smit EF. Predictive role of repair enzymes in the efficacy of cisplatin combinations in pancreatic and lung cancer. Anticancer Res. 2014;34:435–442. [PubMed] [Google Scholar]
- 24.Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026. [DOI] [PubMed] [Google Scholar]
- 25.Guthrie GJ, Charles KA, Roxburgh CS, Horgan PG, McMillan DC, Clarke SJ. The systemic inflammation-based neutrophil-lymphocyte ratio: experience in patients with cancer. Crit Rev Oncol Hematol. 2013;88:218–230. doi: 10.1016/j.critrevonc.2013.03.010. [DOI] [PubMed] [Google Scholar]
- 26.Hohla F, Hopfinger G, Romeder F, Rinnerthaler G, Bezan A, Stattner S, Hauser-Kronberger C, Ulmer H, Greil R. Female gender may predict response to FOLFIRINOX in patients with unresectable pancreatic cancer: a single institution retrospective review. Int J Oncol. 2014;44:319–326. doi: 10.3892/ijo.2013.2176. [DOI] [PubMed] [Google Scholar]
- 27.Basso M, Strippoli A, Orlandi A, Martini M, Calegari MA, Schinzari G, Di Salvatore M, Cenci T, Cassano A, Larocca LM, Barone C. KRAS mutational status affects oxaliplatin-based chemotherapy independently from basal mRNA ERCC-1 expression in metastatic colorectal cancer patients. Br J Cancer. 2013;108:115–120. doi: 10.1038/bjc.2012.526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Spada F, Antonuzzo L, Marconcini R, Radice D, Antonuzzo A, Ricci S, Di Costanzo F, Fontana A, Gelsomino F, Luppi G, Nobili E, Galdy S, Cella CA, Sonzogni A, Pisa E, Barberis M, Fazio N. Oxaliplatin-based chemotherapy in advanced neuroendocrine tumors: clinical outcomes and preliminary correlation with biological factors. Neuroendocrinology. 2016;103:806–814. doi: 10.1159/000444087. [DOI] [PubMed] [Google Scholar]
- 29.Kuwabara K, Kumamoto K, Ishibashi K, Okada N, Ishiguro T, Ohsawa T, Haga N, Miura I, Ishida H. [The relationship between the efficacy of mFOLFOX6 treatment and the expression of TS, DPD, TP, and ERCC-1 in unresectable colorectal cancer] . Gan To Kagaku Ryoho. 2011;38:2224–2227. [PubMed] [Google Scholar]
- 30.Ishibashi K, Okada N, Tajima Y, Ishiguro T, Kuwabara K, Ohsawa T, Kumamoto K, Tsuji Y, Haga N, Iwama T, Ishida H, Onouchi T, Yakabi K. Prediction of the efficacy of modified FOLFOX6 therapy according to the mRNA levels of thymidylate synthase (TS), excision repair cross-complementing-1 and -2 (ERCC-1 and ERCC-2) and methylenetetrahydrofolate dehydrogenase (MTHFD) in the primary lesion of colorectal cancer. Gan To Kagaku Ryoho. 2011;38:2220–2223. [PubMed] [Google Scholar]
- 31.Erkan M, Hausmann S, Michalski CW, Fingerle AA, Dobritz M, Kleeff J, Friess H. The role of stroma in pancreatic cancer: diagnostic and therapeutic implications. Nat Rev Gastroenterol Hepatol. 2012;9:454–467. doi: 10.1038/nrgastro.2012.115. [DOI] [PubMed] [Google Scholar]
- 32.Maithel SK, Coban I, Kneuertz PJ, Kooby DA, El-Rayes BF, Kauh JS, Sarmiento J, Staley CA 3rd, Volkan Adsay N. Differential expression of ERCC1 in pancreas adenocarcinoma: high tumor expression is associated with earlier recurrence and shortened survival after resection. Ann Surg Oncol. 2011;18:2699–2705. doi: 10.1245/s10434-011-1610-x. [DOI] [PubMed] [Google Scholar]
- 33.Bhagwat NR, Roginskaya VY, Acquafondata MB, Dhir R, Wood RD, Niedernhofer LJ. Immunodetection of DNA repair endonuclease ERCC1-XPF in human tissue. Cancer Res. 2009;69:6831–6838. doi: 10.1158/0008-5472.CAN-09-1237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Friboulet L, Olaussen KA, Pignon JP, Shepherd FA, Tsao MS, Graziano S, Kratzke R, Douillard JY, Seymour L, Pirker R, Filipits M, Andre F, Solary E, Ponsonnailles F, Robin A, Stoclin A, Dorvault N, Commo F, Adam J, Vanhecke E, Saulnier P, Thomale J, Le Chevalier T, Dunant A, Rousseau V, Le Teuff G, Brambilla E, Soria JC. ERCC1 isoform expression and DNA repair in non-small-cell lung cancer. N Engl J Med. 2013;368:1101–1110. doi: 10.1056/NEJMoa1214271. [DOI] [PMC free article] [PubMed] [Google Scholar]

