Purpose:

To determine the impact of basal-like and classical subtypes in advanced pancreatic ductal adenocarcinoma (PDAC) and to explore GATA6 expression as a surrogate biomarker.

Experimental Design:

Within the COMPASS trial, patients proceeding to chemotherapy for advanced PDAC undergo tumor biopsy for RNA-sequencing (RNA-seq). Overall response rate (ORR) and overall survival (OS) were stratified by subtypes and according to chemotherapy received. Correlation of GATA6 with the subtypes using gene expression profiling, in situ hybridization (ISH) was explored.

Results:

Between December 2015 and May 2019, 195 patients (95%) had enough tissue for RNA-seq; 39 (20%) were classified as basal-like and 156 (80%) as classical. RECIST response data were available for 157 patients; 29 basal-like and 128 classical where the ORR was 10% versus 33%, respectively (P = 0.02). In patients with basal-like tumors treated with modified FOLFIRINOX (n = 22), the progression rate was 60% compared with 15% in classical PDAC (P = 0.0002). Median OS in the intention-to-treat population (n = 195) was 9.3 months for classical versus 5.9 months for basal-like PDAC (HR, 0.47; 95% confidence interval, 0.32–0.69; P = 0.0001). GATA6 expression by RNA-seq highly correlated with the classifier (P < 0.001) and ISH predicted the subtypes with sensitivity of 89% and specificity of 83%. In a multivariate analysis, GATA6 expression was prognostic (P = 0.02). In exploratory analyses, basal-like tumors, could be identified by keratin 5, were more hypoxic and enriched for a T-cell–inflamed gene expression signature.

Conclusions:

The basal-like subtype is chemoresistant and can be distinguished from classical PDAC by GATA6 expression.

See related commentary by Collisson, p. 4715

Translational Relevance

The transcriptomic basal-like subtype is highly chemoresistant and patients have a shorter median overall survival compared with classical pancreatic ductal adenocarcinoma (PDAC). In this study, survival was lowest in basal-like PDAC treated with modified FOLFIRINOX. GATA6 expression by both RNA-sequencing and in situ hybridization is highly associated with the classifier where low or absent GATA6 is seen in the basal-like subtype.

By 2030, pancreatic ductal adenocarcinoma (PDAC) will become the second leading cause of cancer-related mortality in North America (1). The majority of patients with PDAC present with advanced disease where the mainstay of treatment remains combination chemotherapy. Modified FOLFIRINOX (mFFX) and gemcitabine-nab-paclitaxel (GnP) are the most commonly used regimens resulting in median survival less than 1 year (2, 3). While the need to discover novel approaches is obvious, it is equally important to understand how to select the aforementioned regimens for current patients. There are no randomized data to show superiority of either combination and patient inclusion differences are evident in the two pivotal phase III trials (2, 3). The only molecular predictor of response is prior knowledge of a pathogenic germline variant in a homologous recombination repair gene, which may influence the regimen of choice (4). Other currently targetable genomic variants are uncommon in PDAC.

Gene expression profiling, primarily in resected pancreatic tumors, describes a number of subtypes with considerable overlap, yet presently these do not inform clinical practice (5–7). Collisson and colleagues documented three subtypes (classical, quasimesenchymal, and exocrine-like; ref. 6), Bailey and colleagues four subtypes (immunogenic, progenitor, ADEX, and squamous; ref. 5), and Moffitt and colleagues two subtypes (classical and basal-like; ref. 7). The squamous (Bailey), quasimesenchymal (Collisson), and basal-like (Moffitt) cohorts align well across the classifiers and all three are associated with a poor prognosis in these studies. Despite this, varying tumor cellularity and heterogeneity in clustering methodologies leaves uncertainty as to the most appropriate classifier and furthermore, the clinical application of these subtypes to advanced stage disease is unclear.

In an effort to reconcile and apply existing knowledge, we established the COMPASS trial [Comprehensive Molecular Characterization of Advanced Pancreatic Ductal Adenocarcinomas (PDAC) for Better Treatment Selection: A Prospective Study, NCT NCT02750657]. Unique to this prospective study is the acquisition of tissue prior to chemotherapy in the advanced setting, which then undergoes laser capture microdissection (LCM) to ensure high tumor cellularity. The primary endpoint of feasibility in obtaining a high-quality genome report within 8 weeks in the first 50 patients has been published (8). In this earlier analysis, we determined that a modified Moffitt RNA signature, optimized for use in advanced stage PDAC (classical vs basal-like; Supplementary Fig. S1) may have prognostic impact (8). Furthermore, we found that GATA6, a transcription factor required for normal pancreas development (9), which has been shown to align with the classical subtype, could represent a surrogate marker for classical PDAC (8). Here, we evaluated the modified Moffitt basal-like and classical subtypes together with GATA6 expression on outcomes in patients receiving mFFX or GnP regimens on the expanded COMPASS trial. We further explored specific clinical and pathologic characteristics of the subtypes and evaluated GATA6 as a surrogate biomarker and clinical tool. Post hoc exploratory analyses were performed to seek additional positive biomarkers for the basal-like subtype.

Patient population

The COMPASS trial is a prospective multi-institutional Canadian cohort study. Patient eligibility for the study has been described previously (8). Briefly, patients require a radiologic or histologic diagnosis of locally advanced or metastatic PDAC, suitable for combination chemotherapy, and must consent to a fresh tumor biopsy prior to treatment start. Biopsies can be taken from the primary lesion or any metastatic sites. Patients must not have had prior treatment for advanced disease. Treatment decisions are at the discretion of their medical oncologist. Response to therapy is assessed using CT and measured using RECIST 1.1. Demographics and treatment details, including subsequent treatments, are prospectively collected using an electronic MEDIDATA database. This report includes all patients enrolled from December 2015 until May 2019 and follow-up censored on August 30, 2019. Patients on this study were enrolled at the Princess Margaret Cancer Centre (Toronto, Ontario, Canada), McGill University Health Centre (MUHC, Montreal, Quebec, Canada), and Kingston General Hospital (Kingston, Ontario, Canada) and the study was conducted in accordance with the Declaration of Helsinki. The COMPASS trial has been approved by participating site Institutional Review Board (University Health Network, Toronto, Ontario, Canada; MUHC Centre for Applied Ethics, Montreal, Quebec, Canada; and Queen's University Health Sciences and Affiliated Teaching Hospitals Research Ethics Board, Kingston, Ontario, Canada); each patient provided written informed consent prior to study entry.

RNA-sequencing and GATA6 expression

Frozen biospecimens underwent LCM for tumor enrichment. RNA-sequencing (RNA-seq) analysis was performed at the Ontario Institute of Cancer Research (Ontario, Canada) as described previously (10). Briefly, reads were aligned to the human reference genome (hg38) and transcriptome (Ensembl v84) using STAR v.2.5.2a (11). Duplicated reads were marked using Picard v. 1.121 (https://github.com/broadinstitute/picard). Gene expression was calculated in fragments per kilobase of exon per million reads mapped using the cufflinks package v. 2.2.1 (12). A modified Moffitt classification (classical vs basal-like) was applied to each sample with sufficient RNA for analysis (Supplementary Fig. S1). Cut-off threshold levels for GATA6 expression were determined using the maximal χ2 method on RECIST response and dichotomized GATA6 expression.

GATA6 RNA in situ hybridization

Given our early results, the COMPASS trial was amended (February 1, 2017) to include GATA6 staining using an RNAscope In Situ Hybridization (ISH) Assay (Advanced Cell Diagnostics Inc.). A semiquantitative score was used by the study pathologist (S.E. Fischer; Supplementary Fig. S2A) as reported previously (8). Scoring was applied blinded to results of the modified Moffitt classifier.

IHC analysis of GATA6 and keratins

To provide more widely applicable diagnostic tests for PDAC subtypes, we optimized a protocol for GATA6 IHC (Supplementary Materials and Methods) using a polyclonal anti-GATA6 antibody from R&D (catalog number AF1700), and secondary antibody from Vector (catalog number VECTABA5000). DAB+ (3,3-diaminobenzidine tetrahydrochloride plus, DAKO, catalog number K3468) was used as chromogen and nuclei were counterstained with Mayer hematoxylin (Supplementary Fig. S2B). To assess the pattern of GATA6 staining across larger tumor regions, we used whole sections (n = 30) from a previously described resection cohort with matched RNA-seq data (10) together with biopsies (n = 41) from the advanced cohort.

In an exploratory analysis, we sought additional clinical markers to aid subtype identification; cytokeratins associated with GATA6 expression were identified from RNA-seq data and further explored by IHC (Supplementary Materials and Methods).

Image analysis

To control for potential bias of manual scorings of both ISH or IHC, we performed image analysis on preannotated tumor sections using image analysis software QuPath v0.1.3 (13). Detection parameters were established on unequivocal GATA6-high versus -low versus -absent tumors and confirmed by the study pathologist. Semiquantitative scores were also predicted from image analysis data using the maximal χ2 method.

Statistical analysis

Qualitative variables were compared by Fisher exact test, and quantitative variables by Wilcoxon rank-sum test for pairwise comparison and the Kruskal–Wallis test for multiple group comparison. All patients receiving at least one cycle of chemotherapy were included in the analysis of overall response rate (ORR). Survival curves were plotted using the Kaplan–Meier method and HRs were calculated using Cox proportional hazard regressions with P values calculated using the Wald statistic. All tests were two-sided. Multiple tests P values were adjusted using Benjamini and Hochberg method (14) for independent tests or Benjamini and Yekutieli method (15) for dependent tests, respectively. Statistical significance was set at P = 0.05. All analyses were conducted in R version 3.2 (16). Spearman correlation coefficients were ascertained for evaluating gene expression. Sensitivity, specificity, and accuracy scores were computed to assess prediction quality.

Patient characteristics at baseline

Between December 30, 2015 and May 30,2019, 250 patients were enrolled and 206 were eligible (Fig. 1, consort). Of these, 195 patients (95%) had enough tissue for RNA analysis and are included in this report. Table 1 shows baseline clinical and pathologic characteristics of those patients. Using the modified Moffitt classifier, 39 (20%) baseline tumor samples were basal-like and 156 (80%) classical. Locally advanced disease at diagnosis was present in 24 (12%) and these cases, in this small subset, were all identified as classical (P = 0.005). Liver metastases were present in 97% of basal-like tumors compared with 69% of classical tumors (which includes the locally advanced cases; P < 0.0001). Although basal-like tumors were more frequent in male patients (P = 0.02) the overall sample size was small. Other characteristics were similar between the groups (Table 1).

Figure 1.

Consort diagram of patients enrolled and included on the COMPASS trial. A total of 250 patients were enrolled and 232 patients underwent biopsies. Biopsy sites included liver, pancreas, and peritoneum/omentum. A total of 195 patients were eligible with RNA-seq data representing the study population.

Figure 1.

Consort diagram of patients enrolled and included on the COMPASS trial. A total of 250 patients were enrolled and 232 patients underwent biopsies. Biopsy sites included liver, pancreas, and peritoneum/omentum. A total of 195 patients were eligible with RNA-seq data representing the study population.

Close modal
Table 1.

Baseline characteristics of patients included (n = 195).

Classical (N = 156)Basal-like (N = 39)
N (%)N (%)P
Median age (years) 64.0 (29–84) 65.0 (44–83) 0.75 
Sex 
 Male 83 (53) 29 (74)  
 Female 73 (47) 10 (26) 0.02 
Stage 
 Metastatic 132 (85) 39 (100)  
 Locally advanced 24 (15) 0 (0) 0.005 
Race 
 White 119 (79) 27 (77)  
 Asian 28 (19) 6 (17)  
 African/other 4 (3) 2 (6)  
 Unknown 0.23 
Prior resection 
 Yes 13 (8) 3 (8)  
 No 143 (92) 36 (92) 0.99 
CA19.9 (median, range) 1832 (1–371847) 1124 (1–71956) 0.24 
Ever smoker 
 Yes 80 (51) 23 (59)  
 No 76 (49) 16 (41) 0.47 
Type II DM ≥18 months 
 Yes 32 (21) 8 (21)  
 No 120 (79) 30 (79) 0.99 
Unknown  
Liver metastases 
 Yes 108(69) 38 (97)  
 No 48(31) 1 (3) <0.0001 
HRD genotypea 
 Yes 14 (9) 2 (5)  
 No 142 (91) 37 (95) 0.74 
First chemotherapy 
 mFFX 81 (52) 22 (59)  
GnP regimens 61 (39) 10 (26)  
 GnP alone 43  
 GnP + experimental 18  
 Cisplatin/gem or gem alone 5 (3) 2 (5) 0.25 
 None 9 (6) 5 (13)  
Classical (N = 156)Basal-like (N = 39)
N (%)N (%)P
Median age (years) 64.0 (29–84) 65.0 (44–83) 0.75 
Sex 
 Male 83 (53) 29 (74)  
 Female 73 (47) 10 (26) 0.02 
Stage 
 Metastatic 132 (85) 39 (100)  
 Locally advanced 24 (15) 0 (0) 0.005 
Race 
 White 119 (79) 27 (77)  
 Asian 28 (19) 6 (17)  
 African/other 4 (3) 2 (6)  
 Unknown 0.23 
Prior resection 
 Yes 13 (8) 3 (8)  
 No 143 (92) 36 (92) 0.99 
CA19.9 (median, range) 1832 (1–371847) 1124 (1–71956) 0.24 
Ever smoker 
 Yes 80 (51) 23 (59)  
 No 76 (49) 16 (41) 0.47 
Type II DM ≥18 months 
 Yes 32 (21) 8 (21)  
 No 120 (79) 30 (79) 0.99 
Unknown  
Liver metastases 
 Yes 108(69) 38 (97)  
 No 48(31) 1 (3) <0.0001 
HRD genotypea 
 Yes 14 (9) 2 (5)  
 No 142 (91) 37 (95) 0.74 
First chemotherapy 
 mFFX 81 (52) 22 (59)  
GnP regimens 61 (39) 10 (26)  
 GnP alone 43  
 GnP + experimental 18  
 Cisplatin/gem or gem alone 5 (3) 2 (5) 0.25 
 None 9 (6) 5 (13)  

Note: Baseline characteristics of cases enrolled according to modified Moffitt classification (classical vs basal-like).

Abbreviations: DM, diabetes mellitus; gem, gemcitabine; HRD, homologous recombination deficiency.

aIncludes both somatic and germline HRD cases.

Response to chemotherapy according to modified Moffitt classification.

Of the 195 patients, 14 (7%) did not receive any chemotherapy and were considered nonevaluable (NE). A further 23 patients (12%) died as a result of rapid functional decline prior to their first scan, of which 19 received only one cycle of chemotherapy; five of these 23 had basal-like PDAC. One patient receiving mFFX did not have measurable disease at enrollment. Accordingly, RECIST response data were available for 157 patients (81%) including 29 patients with basal-like tumors and 128 with classical tumors (Fig. 2A). The ORR in classical PDAC was 33% versus 10% in basal-like PDAC (P = 0.02). The rates of progression by RECIST criteria at first CT image were much higher in basal-like versus classical PDAC (52% vs 16%; P < 0.0001). Figure 2A shows the percentage change in target lesions, demonstrating chemoresistance of the basal-like subtype. In patients treated with mFFX and evaluable for response (n = 91), progression was evident in 60% of basal-like versus 15% of classical PDAC (P = 0.0002; Fig. 2B). The ORR was 29.6% versus 10% in classical versus basal-like PDAC (P = 0.09). One patient in the latter group with a partial response had a germline BRCA2 pathogenic variant and displayed genomic characteristics of homologous recombination deficiency. The numbers treated with GnP regimens and available for response were small, progression of disease was seen in three of nine (33%) patients with basal-like versus eight of 54 (15%) with classical tumors (P = 0.18; Fig. 2C). The ORR was 39% versus 11% in classical versus basal-like PDAC, respectively (P = 0.14). Of note, 20 of 63 (32%) received additional experimental agents in this group.

Figure 2.

Waterfall plots demonstrating tumor size change according to modified Moffitt classifier. A, Tumor size change in all patients included (n = 194*): This includes any chemotherapy received. The nonevaluable patients did not have imaging to determine response. B, Tumor size change in patients receiving first-line mFFX (n = 102*). C, Tumor size change in patients receiving GnP regimens (n = 71). *, one patient with nonmeasurable disease is not included; ×, new lesions.

Figure 2.

Waterfall plots demonstrating tumor size change according to modified Moffitt classifier. A, Tumor size change in all patients included (n = 194*): This includes any chemotherapy received. The nonevaluable patients did not have imaging to determine response. B, Tumor size change in patients receiving first-line mFFX (n = 102*). C, Tumor size change in patients receiving GnP regimens (n = 71). *, one patient with nonmeasurable disease is not included; ×, new lesions.

Close modal

Overall survival according to the modified Moffitt classification

Overall survival (OS) in the intention-to-treat population (n = 195) is shown in Fig. 3A. Median follow-up was 7.17 months. Median OS according to receipt of chemotherapy is shown in Fig. 3B. In patients receiving mFFX (n = 103), where performance status was less likely to confound results, median OS was 6.5 months in basal-like versus 10.6 months in classical subgroups [HR, 0.33; 95% confidence interval (CI), 0.19–0.60; P = 0.0001). These observations suggest favorable impact of mFFX in classical PDAC but little impact of mFFX in the basal-like population (Fig. 3C). In contrast, there was no difference between subgroups when treated with GnP regimens, where median OS, was 8.12 months in basal-like versus 8.19 months in classical groups, respectively (HR, 0.80; 95% CI, 0.40–1.60; P = 0.53; Fig. 3D). In a multivariate Cox proportional hazard regression analysis, the Moffitt subtype remained highly prognostic (P = 0.018). Substituting GATA6 expression for the Moffit subtype also demonstrated the prognostic impact of GATA6 in the model, again supporting its use as a biomarker of the subtypes. Of note, stage (locally advanced vs metastatic) or chemotherapy type had no impact in this observational cohort study (Supplementary Fig. S3). To further explore whether there was a significant interaction between mFFX or GnP and the subtypes, we performed an interaction analysis. There was no statistically significant difference to suggest one chemotherapy regimen for one particular subtype, although basal-like tumors trended toward improved survival with GnP (P = 0.08). Of note, the modified Moffitt classifier used in this study outperforms the previously published Moffitt classifier in identifying the poor prognostic basal-like subtype (Supplementary Fig. S1B).

Figure 3.

Kaplan–Meier OS curves according to modified Moffitt subtype and chemotherapy received. A, OS in the intention-to-treat population (n = 195) which includes patients who did not receive chemotherapy or who were NE. B, OS in patients receiving first-line mFFX or GnP regimens (at least one cycle) and is presented according to modified Moffitt subtype (n = 174). This graph integrates curves in C and D. C, OS in patients receiving ≥ 1 cycle mFFX (n = 103) according to modified Moffitt subtype. D, OS in patients receiving ≥ 1 cycle GnP regimens (n = 71) according to modified Moffitt subtype.

Figure 3.

Kaplan–Meier OS curves according to modified Moffitt subtype and chemotherapy received. A, OS in the intention-to-treat population (n = 195) which includes patients who did not receive chemotherapy or who were NE. B, OS in patients receiving first-line mFFX or GnP regimens (at least one cycle) and is presented according to modified Moffitt subtype (n = 174). This graph integrates curves in C and D. C, OS in patients receiving ≥ 1 cycle mFFX (n = 103) according to modified Moffitt subtype. D, OS in patients receiving ≥ 1 cycle GnP regimens (n = 71) according to modified Moffitt subtype.

Close modal

GATA6 expression by RNA-seq and RNA ISH is associated with modified Moffitt subtypes.

GATA6 expression remained strongly associated with the modified Moffitt transcriptomic classifier (P < 0.001) in this expanded cohort (Fig. 4A, left). In addition, the proposed RNA ISH semiquantitative score was highly associated with GATA6 gene expression (RNA-seq; P < 0.001; Fig. 4A, right). Matched RNA-seq and ISH results were available in 106 patients (23 with basal-like and 83 with classical subtypes). Semiquantitative scoring of GATA6 ISH confirmed higher GATA6 expression (74/83 score 2–4) in classical versus basal-like PDAC (19/23 score 0–1). Furthermore, GATA6 ISH correlated with modified Moffitt with a sensitivity of 89%, specificity of 83%, and accuracy of 88%. Both manual scores and modified Moffitt calls could be predicted from image analysis data with concordance of 92% and 81%, respectively, confirming reproducibility of semiquantitative assessment (n = 106). The modified Moffitt signature remained prognostic in this staining subcohort (Fig. 4B); both GATA6 ISH SQ scoring (Fig. 4C) and subtyping inferred from image analysis of GATA6 ISH (Fig. 4D) predicted outcome in a similar manner.

Figure 4.

GATA6 expression is associated with modified Moffitt subtypes in advanced PDAC. A, Gata6 expression by RNA-seq versus modified Moffitt subtypes (left), and GATA6 expression by RNA-seq versus GATA6 ISH (right). Scores of 0–1 reflect the basal-like subtype and 2–5 the classical subtype. B, Kaplan–Meier curve of OS by modified Moffitt in patients with matched tissue for RNA-seq and GATA6 ISH analysis (n = 106). C, Kaplan–Meier curves of OS by GATA6 ISH semiquantitative analysis in patients with matched tissue for RNA-seq and GATA6 ISH analysis (n = 106). D, Kaplan–Meier curve of GATA6 by QuPath image analysis in those patients with matched tissue for RNA-seq and GATA6 ISH (n = 106).

Figure 4.

GATA6 expression is associated with modified Moffitt subtypes in advanced PDAC. A, Gata6 expression by RNA-seq versus modified Moffitt subtypes (left), and GATA6 expression by RNA-seq versus GATA6 ISH (right). Scores of 0–1 reflect the basal-like subtype and 2–5 the classical subtype. B, Kaplan–Meier curve of OS by modified Moffitt in patients with matched tissue for RNA-seq and GATA6 ISH analysis (n = 106). C, Kaplan–Meier curves of OS by GATA6 ISH semiquantitative analysis in patients with matched tissue for RNA-seq and GATA6 ISH analysis (n = 106). D, Kaplan–Meier curve of GATA6 by QuPath image analysis in those patients with matched tissue for RNA-seq and GATA6 ISH (n = 106).

Close modal

GATA6 IHC may discriminate basal-like from classical PDAC

Matched IHC and ISH results were available in only 78 advanced PDAC cases. GATA6 levels by IHC and ISH were well correlated using quantitative assessment (Supplementary Fig. S4A) and semiquantitative scoring (concordance 88%), indicating that GATA6 protein levels mirror RNA expression and could aid subtype identification when RNA detection is not feasible. Indeed, IHC-based semiquantitative scoring identified most patients with classical subtype tumors by strong and moderate GATA6 staining (52/63 with scores 2–4), while basal-like subtype patients mostly exhibited no or weak GATA6 staining (9/15 with scores 0–1), so that GATA6 protein detection by IHC was associated with modified Moffitt subtypes in advanced PDAC with a sensitivity of 83%, specificity of 60%, and accuracy of 78%. Once more, this was confirmed by quantitative assessment (Supplementary Fig. S4B) and the concordance between prediction of GATA6 scoring from image analysis to manual scoring of GATA6 was 90%.

Tissue distribution of GATA6 by IHC in a subset or resectable and metastatic PDAC

Recent data are emerging that basal and classical subtypes can coexist in PDAC (17, 18). We therefore explored potential variation in GATA6 expression patterns. We used whole sections from selected resected cases (n = 30) in addition to needle biopsies (n = 41). Although early stage tumors may not necessarily reflect the biology of advanced disease, adequate tumor content is available for more complete evaluation. GATA6 staining (IHC) in resected specimens that were basal-like (n = 14) and classical (n = 16) also associated with the Moffitt subtypes: extensive immunopositivity for GATA6 (>50% of tumor cells with score 2 or higher) was found in nine of 16 (56%) classical tumors versus one of 14 (7%) basal-like tumors (Supplementary Table S1). Interestingly, variable GATA6 immunopositivity (<50% of tumor cells with score 2 or higher) was present in four of 16 (25%) and four of 14 (28%) of classical and basal-like tumors, respectively, documenting a group where these subtypes may coexist. This was furthermore observed in a number of advanced PDAC biopsies, which also exhibited variable GATA6 expression by ISH and IHC (Supplementary Fig. S5), demonstrating that regional GATA6 heterogeneity can exist in resectable and advanced-stage tumors. These differences were also observed at the cellular level by image analysis (Supplementary Fig. S6). In sum, GATA6 staining patterns were widely comparable across whole sections of 22 of 30 (73%) resection cases. Variable GATA6 immunopositivity was present in a subset of both, classical and basal-like subtypes, in resectable and advanced disease, which may point at the presence of classical and basal regions in the same tumor.

Keratin 5 may positively identify the basal-like subtype

GATA6 positively identifies classical PDAC, but markers for the basal subtype are lacking.

In an exploratory analysis, we evaluated keratin markers associated with GATA6 expression. In-line with their use as basal markers in other tumor types (19, 20), keratins 15, 5/6, 23, and 14 were inversely correlated with GATA6 expression and thus the classical subtype (Supplementary Fig. S7A). In this post hoc analysis, none of the identified cytokeratins were superior to GATA6 in their association with modified Moffitt subgroups, including keratin 17, a prognostic marker in PDAC (ref. 21; Supplementary Fig. S7B). Among these, keratin 5 (CK5) demonstrated the strongest expression differences between basal-like and classical tumors and was found to be complementary to GATA6 expression in our cohort (Supplementary 8). Furthermore, GATA6 and keratin 5 often demonstrated complementary staining pattern by IHC in PDAC tissues, including in 41 COMPASS biopsies and 30 resected PDAC whole sections (Fig. 5; Supplementary Table S1). From these specimens, we observed the presence of both GATA6 and CK5 staining in a subset of cases (Fig. 5, bottom). Indeed, the intratumoral staining pattern of the two markers was predominantly inversely correlated in 149 individual tumor regions from the 30 resected cases (Fig. 6A). Of note, this analysis revealed a small number of regions that contained considerable number of both CK5+ and GATA6+ cells (Fig. 6A). Double immunostaining confirmed distinct GATA6+/CK5 and GATA6/CK5+ regions within the same tumor (Fig. 6B) and in individual ducts (Fig. 6C), which further support the notion that basal-like and classical programs can coexist in the same tumor. Overall, many basal-like cases of advanced PDAC showed CK5 positivity (10/19, 53%), whereas most classical tumors (22/23, 96%) exhibited scant (<10%) or negative CK5 staining. Keratin 5 was thus highly specific and also showed remarkable intratumoral complementarity to GATA6 staining suggesting a clinically relevant biomarker of the basal-like subtype.

Figure 5.

Pathology images comparing GATA6 staining by ISH and IHC, together with CK5 IHC staining. A, Advanced PDAC cases: COMP-022: classic with glandular architecture [hematoxylin and eosin (H&E)], GATA6 ISH score 3, GATA6 IHC score 2, and CK5-negative (rare positive cells); magnification, 100×. COMPA-0234: basal-like with squamous features (H&E), GATA6 ISH score 1, GATA6 IHC score 1 (weak/focal), and CK5 positive; magnification, 100×. COMP-0135: basal-like with poor differentiation (H&E), GATA6 ISH score 2 (variable distribution), GATA6 IHC score 2 (variable distribution), and CK5 positive; magnification, 100×. B, Resected PDAC cases: expression pattern of GATA6 and CK5 in resected PDAC. PCSI_639: classic with glandular architecture (H&E), GATA6 ISH score 3, GATA6 IHC score 2, and CK5 negative; magnification, 100×. PCSI_588: basal-like with squamous features (H&E), GATA6 IHC score 1 (weak/focal), and CK5 positive; magnification, 100×. PCSI_645: classic with dual phenotype (glandular and squamous) on HE, GATA6 IHC score 2 (variable distribution), and CK5 positive; magnification, 25×.

Figure 5.

Pathology images comparing GATA6 staining by ISH and IHC, together with CK5 IHC staining. A, Advanced PDAC cases: COMP-022: classic with glandular architecture [hematoxylin and eosin (H&E)], GATA6 ISH score 3, GATA6 IHC score 2, and CK5-negative (rare positive cells); magnification, 100×. COMPA-0234: basal-like with squamous features (H&E), GATA6 ISH score 1, GATA6 IHC score 1 (weak/focal), and CK5 positive; magnification, 100×. COMP-0135: basal-like with poor differentiation (H&E), GATA6 ISH score 2 (variable distribution), GATA6 IHC score 2 (variable distribution), and CK5 positive; magnification, 100×. B, Resected PDAC cases: expression pattern of GATA6 and CK5 in resected PDAC. PCSI_639: classic with glandular architecture (H&E), GATA6 ISH score 3, GATA6 IHC score 2, and CK5 negative; magnification, 100×. PCSI_588: basal-like with squamous features (H&E), GATA6 IHC score 1 (weak/focal), and CK5 positive; magnification, 100×. PCSI_645: classic with dual phenotype (glandular and squamous) on HE, GATA6 IHC score 2 (variable distribution), and CK5 positive; magnification, 25×.

Close modal
Figure 6.

Tissue pattern of GATA6 and keratin 5 expression. A, IHC staining of GATA6 and keratin 5 on serial sections from resected PDAC specimen (n = 30). Representative images, magnification, 25× (left). Quantification of the percentage of GATA6+ or CK5+ cells, respectively, in 149 matched regions on adjacent sections (right). B, Dual immunostaining of GATA6 (brown) or CK5 (magenta) in resected PDAC revealing distinct regions of GATA6 or CK5 immunoreactivity, magnification, 25n×. C, Dual immunostaining of GATA6 (brown) or CK5 (magenta) revealing GATA6 and CK5 immunoreactivity in the same tumor ducts. Resected PDAC (left); advanced PDAC (right); magnification, 400×.

Figure 6.

Tissue pattern of GATA6 and keratin 5 expression. A, IHC staining of GATA6 and keratin 5 on serial sections from resected PDAC specimen (n = 30). Representative images, magnification, 25× (left). Quantification of the percentage of GATA6+ or CK5+ cells, respectively, in 149 matched regions on adjacent sections (right). B, Dual immunostaining of GATA6 (brown) or CK5 (magenta) in resected PDAC revealing distinct regions of GATA6 or CK5 immunoreactivity, magnification, 25n×. C, Dual immunostaining of GATA6 (brown) or CK5 (magenta) revealing GATA6 and CK5 immunoreactivity in the same tumor ducts. Resected PDAC (left); advanced PDAC (right); magnification, 400×.

Close modal

Additional molecular characteristics of the basal-like phenotype

Among the 195 eligible COMPASS patients, all eight (4%) with adenosquamous histology were basal-like and stained positive for keratin 5 by IHC, with negligible GATA6 expression by RNA ISH. We have previously shown that the basal-like subgroup is enriched in a hypoxia-associated gene signature by gene set enrichment analysis (22) and this observation is retained in this expanded dataset (P = 0.0003). In addition, we found higher PD-L1 expression in the basal-like cohort (P < 0.001), higher PD-1 expression (P < 0.001), and enrichment of a T-cell–inflamed signature reported previously (refs. 23, 24; P = 0.007; Supplementary Fig. S9). Tumor mutational burden was not different between groups (2.02 mutations/Mb vs 1.96 mutations/Mb) and was consistent with that seen in an unselected PDAC cohort (25).

Combination chemotherapy is used in the treatment of most patients with advanced PDAC, yet the field is lacking robust biomarkers of outcome to guide regimen selection. Here, we show that patients with tumors of a modified “basal-like” phenotype, or those with low GATA6 RNA expression, have inferior outcomes compared with those with the “classical” phenotype. The latter are accurately identified by high GATA6 expression and positive GATA6 staining by ISH. Our data also suggests that basal-like tumors are particularly resistant to mFFX, warranting further investigation.

Both the PRODIGE4/ACCORD 11 and the MPACT PDAC trials of metastatic disease demonstrated an improvement in survival with FOLFIRINOX and GnP across all subcohorts compared with gemcitabine alone (2, 3), yet they provide little insight into which subgroups might benefit the most. Notably, our study shows no superiority in either regimen in an unselected population with regard to survival. In the aforementioned trials, histologic groups were not documented in either study, which is not unusual because many patients have a diagnosis made from very small samples or brushings. In contrast, the histologic classification in resected specimens can be more easily reported and the PRODIGE24/CCTG PA6 trial of mFFX in the adjuvant setting documented the prognostic impact of tumor grade in multivariate analysis (26) In patients receiving mFFX, those with well-differentiated tumors benefited the most (HR, 0.52; 95% CI, 0.34–0.81), whereas the impact in poorly differentiated tumors was not significant. Although limitations to the three-tiered histologic classification (poor, moderate, and well-differentiated) in PDAC have been noted (27), well-differentiated tumors highly express the classical program and GATA6 (28).

The resistance of the basal-like subtype to mFFX is supported by a recent collaborative study by Tiriac and colleagues (29) demonstrating that patient-derived organoid chemotherapy signatures may predict treatment response. The signatures indicative of individual cytotoxic agents were applied to our COMPASS cohort suggesting that the basal-like cohort subgroup was most likely to have a nonoxaliplatin-sensitive signature (29). We furthermore hypothesize that basal-like tumors may have limited sensitivity to 5-fluorouracil (5-FU). Martinelli and colleagues demonstrated GATA6 loss in resected PDAC with a basal-like phenotype in the ESPAC-3 trial, and shorter survival in these patients when treated with adjuvant 5-FU. This study also showed that GATA6-low cell lines derived from patient-derived xenografts were particularly resistant to 5-FU but not gemcitabine (30). Notably oxaliplatin was not evaluated. In search of treatment alternatives, we report here that basal-like tumors had higher hypoxia scores, and higher PD-1 and PD-L1 expression with enrichment of a T-cell–inflamed signature (24), which may be predictive of immunotherapy 6 in this chemoresistant group. Similarly, triple-negative breast cancers, although associated with worse outcomes, have higher levels of tumor-infiltrating lymphocytes compared with hormone receptor–positive, HER2 tumors. The impact of immune populations within subtypes in PDAC will require further investigation (31).

Clinical applicability of RNA-seq and tumor enrichment by LCM is currently limited given tissue acquisition, cost, and time to reporting. GATA6 detection from formalin-fixed, paraffin-embedded needle biopsies at diagnosis is therefore an attractive surrogate for transcriptomic classifiers. We demonstrate concordance of GATA6 ISH with the subtypes with sensitivity and specificity of over 80% in our tumor-enriched samples. Of note, the GATA6 gene is not part of the original Moffitt subtype signatures but rather the Bailey squamous classifier, which largely overlaps with Moffitt calls in high purity samples (5). The number of tissue specimens available for matched ISH, IHC, and RNA-seq was low in our study (n = 78, 40%). Therefore, although specificity was much lower for IHC compared with ISH, a prospective study with adequate tissue for matched analysis is needed. Recognizing that the identification of the basal-like subtype is critical and that GATA6 is a negative marker, we sought additional positive keratin biomarkers that may be more feasible for the practicing clinician. Of these, keratin 5 predicted outcomes best after GATA6 expression and was found to exhibit high complementarity to GATA6 staining pattern and RNA expression levels. Moreover, combined keratin 5 and GATA6 stainings on serial sections and by double immune-staining have consistently suggested that basal-like and classical elements can coexist in a subset of PDAC cases, which strongly reinforces the need for a positive basal-like biomarker and has major implications for rationalizing subtype-specific treatments. We are currently evaluating combined staining of GATA6 and keratin 5 on the COMPASS trial.

Notably microdissected tissue, although impractical in laboratory medicine practice, most accurately detects tumor gene expression, with comparatively less exocrine and immune compartments compared with TCGA datasets, as recently described (32). This therefore implies that more reliable biomarkers can be determined from highly cellular specimens. CA-19.9 is the only approved biomarker for monitoring disease in the advanced setting (33) and the POLO trial has now documented a benefit for maintaining PARPi in patients with germline BRCA mutations (4). Robust subtyping of pancreatic cancer will be critical to advancing the field, GATA6 as a single biomarker and highly correlated with the Moffitt classifier will now be evaluated in a prospective trial.

This study is limited by few progression biopsies to understand the stability of the subtypes under selective pressure during chemotherapy. This is especially interesting in light of the coexistence of basal-like and classical elements, documented here and elsewhere (17, 18). In addition, the numbers of basal-like tumors treated with GnP regimens is low and the GnP group is potentially confounded by performance status. The interaction term for chemotherapy type and subtype was not significant in this study although numbers were low. We therefore cannot conclude whether GnP is a better strategy in the basal-like cohort, rather our data suggests alternative therapies are urgently needed, and clinical trials to evaluate this particular group are warranted. With mFFX as current treatment of choice in the adjuvant setting, understanding chemotherapy response to subtypes has increasing importance. It should also be noted that the response rates and survival between those receiving mFFX and GnP were not statistically different in this analysis. This is supported by the recent HALO trial 109–321 study where response rates and OS are comparable with historic outcomes with mFFX (2, 34). This furthermore supports the need to understand which populations can benefit most from these regimens and a prospective trial has now been planned.

In the major tumor types of lung and colorectal cancer, factors such as histologic subtype, molecular profile, and PD-L1 status can influence the choice of upfront systemic treatment in advanced disease and have resulted in survival gains (35–37). Because PDAC will soon become the second leading cause of cancer-related mortality, it behooves the oncology community to invest in biomarkers helpful for selecting standard chemotherapy. In this study, we confirm the prognostic impact of the modified Moffitt subtypes and demonstrate that basal-like PDAC responds poorly to mFFX. The basal-like cohort can be accurately identified by GATA6 RNA expression, providing a putative single important biomarker in clinical trial design.

G.M. O’Kane reports receiving speakers bureau honoraria from Roche and Eisai. J.M.S. Bartlett reports receiving commercial research grants from Thermo Fisher Scientific, Genoptix, Agendia, NanoString Technologies, Inc., Stratifyer GmbH, and Biotheranostics, Inc., speakers bureau honoraria from NanoString Technologies, Inc., Oncology Education, and Biotheranostics, Inc., and is an advisory board member/unpaid consultant for Insight Genetics, Inc., BioNTech AG, Biotheranostics, Inc., Pfizer, Rna Diagnostics, and oncoXchange. J.J. Knox is an employee/paid consultant for Merck, and reports receiving commercial research grants from Astra Zeneca, Ibsen, Roche, and Merck. No potential conflicts of interest were disclosed by the other authors.

Conception and design: G.M. O'Kane, M. Masoomian, J.J. Knox, S.E. Fischer

Development of methodology: G.M. O'Kane, B.T. Grünwald, J.J. Knox, S.E. Fischer

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G.M. O'Kane, B.T. Grünwald, G.-H. Jang, M. Masoomian, S. Picardo, Y. Wang, J.K. Miller, B. Lam, P.M. Krzyzanowski, I.M. Lungu, J.M.S. Bartlett, F. Vyas, R. Khokha, J. Biagi, D. Chadwick, S. Ramotar, S. Hutchinson, A. Dodd, J.M. Wilson, G. Zogopoulos, S. Gallinger, J.J. Knox, S.E. Fischer

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G.M. O'Kane, B.T. Grünwald, G.-H. Jang, R.C. Grant, R.E. Denroche, Y. Wang, F. Vyas, F. Notta, S. Gallinger, J.J. Knox

Writing, review, and/or revision of the manuscript: G.M. O'Kane, B.T. Grünwald, G.-H. Jang, R.C. Grant, Y. Wang, J.K. Miller, J. Biagi, G. Zogopoulos, S. Gallinger, J.J. Knox, S.E. Fischer

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): G.M. O'Kane, S. Picardo, R.E. Denroche, A. Zhang, J.K. Miller, I.M. Lungu, J.M.S. Bartlett, M. Peralta, R. Khokha, S. Ramotar, S. Hutchinson, A. Dodd, J.M. Wilson, G. Zogopoulos, S. Gallinger, J.J. Knox, S.E. Fischer

Study supervision: G.M. O'Kane, P.M. Krzyzanowski, J.J. Knox, S.E. Fischer

Other (collection of data): M. Masoomian

Other (primary study coordinator): S. Ramotar

This study was conducted with support of the Ontario Institute for Cancer Research (PanCuRx Translational Research Initiative) through funding provided by the Government of Ontario, the Wallace McCain Centre for Pancreatic Cancer supported by the Princess Margaret Cancer Foundation, the Terry Fox Research Institute, the Canadian Cancer Society Research Institute, and the Pancreatic Cancer Canada Foundation. The study was also supported by charitable donations from the Canadian Friends of the Hebrew University (Alex U. Soyka). J.J. Knox is the recipient of the Lewitt Chair in Pancreatic Cancer. G.M. O'Kane is supported by the Lewitt fellowship. S. Gallinger is the recipient of an Investigator Award from OICR. G. Zogopoulos is a clinical research scholar of the Fonds de recherche du Québec–Santé. B.T. Grünwald was supported by the Princess Margaret Cancer Foundation, EMBO (ALTF 116-2018), and the Alexander-von-Humboldt Foundation (DEU 1199182 FLF-P). We acknowledge the contributions of team members at OICR within the Diagnostic Development platform and the Genomics & Bioinformatics platform (genomics.oicr.on.ca).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
.
Cancer statistics, 2016
.
CA Cancer J Clin
2016
;
66
:
7
30
.
2.
Conroy
T
,
Desseigne
F
,
Ychou
M
,
Bouche
O
,
Guimbaud
R
,
Becouarn
Y
, et al
.
FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer
.
N Engl J Med
2011
;
364
:
1817
25
.
3.
Von Hoff
DD
,
Ervin
T
,
Arena
FP
,
Chiorean
EG
,
Infante
J
,
Moore
M
, et al
.
Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine
.
N Engl J Med
2013
;
369
:
1691
703
.
4.
Golan
T
,
Hammel
P
,
Reni
M
,
Van Cutsem
E
,
Macarulla
T
,
Hall
MJ
, et al
.
Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer
.
N Engl J Med
2019
;
381
:
317
27
.
5.
Bailey
P
,
Chang
DK
,
Nones
K
,
Johns
AL
,
Patch
AM
,
Gingras
MC
, et al
.
Genomic analyses identify molecular subtypes of pancreatic cancer
.
Nature
2016
;
531
:
47
52
.
6.
Collisson
EA
,
Sadanandam
A
,
Olson
P
,
Gibb
WJ
,
Truitt
M
,
Gu
S
, et al
.
Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy
.
Nat Med
2011
;
17
:
500
3
.
7.
Moffitt
RA
,
Marayati
R
,
Flate
EL
,
Volmar
KE
,
Loeza
SG
,
Hoadley
KA
, et al
.
Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma
.
Nat Genet
2015
;
47
:
1168
78
.
8.
Aung
KL
,
Fischer
SE
,
Denroche
RE
,
Jang
GH
,
Dodd
A
,
Creighton
S
, et al
.
Genomics-driven precision medicine for advanced pancreatic cancer: early results from the COMPASS trial
.
Clin Cancer Res
2018
;
24
:
1344
54
.
9.
Shi
ZD
,
Lee
K
,
Yang
D
,
Amin
S
,
Verma
N
,
Li
QV
, et al
.
Genome editing in hPSCs reveals GATA6 haploinsufficiency and a genetic interaction with GATA4 in human pancreatic development
.
Cell Stem Cell
2017
;
20
:
675
88
.
10.
Connor
AA
,
Denroche
RE
,
Jang
GH
,
Timms
L
,
Kalimuthu
SN
,
Selander
I
, et al
.
Association of distinct mutational signatures with correlates of increased immune activity in pancreatic ductal adenocarcinoma
.
JAMA Oncol
2017
;
3
:
774
83
.
11.
Dobin
A
,
Davis
CA
,
Schlesinger
F
,
Drenkow
J
,
Zaleski
C
,
Jha
S
, et al
.
STAR: ultrafast universal RNA-seq aligner
.
Bioinformatics
2013
;
29
:
15
21
.
12.
Trapnell
C
,
Williams
BA
,
Pertea
G
,
Mortazavi
A
,
Kwan
G
,
van Baren
MJ
, et al
.
Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation
.
Nat Biotechnol
2010
;
28
:
511
5
.
13.
Bankhead
P
,
Loughrey
MB
,
Fernandez
JA
,
Dombrowski
Y
,
McArt
DG
,
Dunne
PD
, et al
.
QuPath: open source software for digital pathology image analysis
.
Sci Rep
2017
;
7
:
16878
.
14.
Benjamini
Y
,
Hochberg
Y
.
Controlling the false discovery rate: a practical and powerful approach to multiple testing
.
J R Stat Soc Series B
1995
;
57
:
289
300
.
15.
Benjamini
Y
,
Yekutieli
D
.
The control of the false discovery rate in multiple testing under dependency
.
Ann Stat
2001
;
29
:
1165
1188
.
16.
R Core Team
.
R: a language and environment for statistical computing
.
2015
. Available from: https://www.R-project.org/.
17.
Chan-Seng-Yue
M
,
Kim
JC
,
Wilson
GW
,
Ng
K
,
Figueroa
EF
,
O'Kane
GM
, et al
.
Transcription phenotypes of pancreatic cancer are driven by genomic events during tumor evolution
.
Nat Genet
2020
;
52
:
231
40
.
18.
Hayashi A
FJ
,
Chen
R
,
Ho
YJ
,
Makohon-Moore
AP
,
Lecomte
N
,
Zhong
Y
, et al
.
A unifying paradigm for transcriptional heterogeneity and squamous features in pancreatic ductal adenocarcinoma
.
Nat Cancer
2020
;
1
:
59
74
.
19.
Abd El-Rehim
DM
,
Pinder
SE
,
Paish
CE
,
Bell
J
,
Blamey
RW
,
Robertson
JF
, et al
.
Expression of luminal and basal cytokeratins in human breast carcinoma
.
J Pathol
2004
;
203
:
661
71
.
20.
Khanom
R
,
Sakamoto
K
,
Pal
SK
,
Shimada
Y
,
Morita
K
,
Omura
K
, et al
.
Expression of basal cell keratin 15 and keratin 19 in oral squamous neoplasms represents diverse pathophysiologies
.
Histol Histopathol
2012
;
27
:
949
59
.
21.
Roa-Pena
L
,
Leiton
CV
,
Babu
S
,
Pan
CH
,
Vanner
EA
,
Akalin
A
, et al
.
Keratin 17 identifies the most lethal molecular subtype of pancreatic cancer
.
Sci Rep
2019
;
9
:
11239
.
22.
Connor
AA
,
Denroche
RE
,
Jang
GH
,
Lemire
M
,
Zhang
A
,
Chan-Seng-Yue
M
, et al
.
Integration of genomic and transcriptional features in pancreatic cancer reveals increased cell cycle progression in metastases
.
Cancer Cell
2019
;
35
:
267
82
.
23.
Trujillo
JA
,
Sweis
RF
,
Bao
R
,
Luke
JJ
.
T cell-inflamed versus non-T cell-inflamed tumors: a conceptual framework for cancer immunotherapy drug development and combination therapy selection
.
Cancer Immunol Res
2018
;
6
:
990
1000
.
24.
Spranger
S
,
Bao
R
,
Gajewski
TF
.
Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity
.
Nature
2015
;
523
:
231
5
.
25.
Cancer Genome Atlas Research Network
.
Integrated genomic characterization of pancreatic ductal adenocarcinoma
.
Cancer Cell
2017
;
32
:
185
203
.
26.
Conroy
T
,
Hammel
P
,
Hebbar
M
,
Ben Abdelghani
M
,
Wei
AC
,
Raoul
JL
, et al
.
FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer
.
N Engl J Med
2018
;
379
:
2395
406
.
27.
N Kalimuthu
S
,
Wilson
GW
,
Grant
RC
,
Seto
M
,
O'Kane
G
,
Vajpeyi
R
, et al
.
Morphological classification of pancreatic ductal adenocarcinoma that predicts molecular subtypes and correlates with clinical outcome
.
Gut
2020
;
69
:
317
28
.
28.
Collisson
EA
,
Bailey
P
,
Chang
DK
,
Biankin
AV
.
Molecular subtypes of pancreatic cancer
.
Nat Rev Gastroenterol Hepatol
2019
;
16
:
207
20
.
29.
Tiriac
H
,
Belleau
P
,
Engle
DD
,
Plenker
D
,
Deschenes
A
,
Somerville
T
, et al
.
Organoid profiling identifies common responders to chemotherapy in pancreatic cancer
.
Cancer Discov
2018
;
8
:
1112
29
.
30.
Martinelli
P
,
Carrillo-de Santa Pau
E
,
Cox
T
,
Sainz
B
Jr
,
Dusetti
N
,
Greenhalf
W
, et al
.
GATA6 regulates EMT and tumour dissemination, and is a marker of response to adjuvant chemotherapy in pancreatic cancer
.
Gut
2017
;
66
:
1665
76
.
31.
Stanton
SE
,
Adams
S
,
Disis
ML
.
Variation in the incidence and magnitude of tumor-infiltrating lymphocytes in breast cancer subtypes: a systematic review
.
JAMA Oncol
2016
;
2
:
1354
60
.
32.
Peng
XL
,
Moffitt
RA
,
Torphy
RJ
,
Volmar
KE
,
Yeh
JJ
.
De novo compartment deconvolution and weight estimation of tumor samples using DECODER
.
Nat Commun
2019
;
10
:
4729
.
33.
Winter
JM
,
Yeo
CJ
,
Brody
JR
.
Diagnostic, prognostic, and predictive biomarkers in pancreatic cancer
.
J Surg Oncol
2013
;
107
:
15
22
.
34.
Tempero
MA
,
Cutsem
EV
,
Sigal
D
,
Oh
DY
,
Fazio
N
,
Macarulla
T
,
Hitre
E
, et al
.
HALO 109–301: a randomized, double-blind, placebo-controlled, phase 3 study of pegvorhyaluronidase alfa (PEGPH20)+ nab-paclitaxel/gemcitabine (AG) in patients (pts) with previously untreated hyaluronan (HA)-high metastatic pancreatic ductal adenocarcinoma (mPDA)
.
J Clin Oncol 2020 Feb 4
[Epub ahead of print].
35.
Scagliotti
GV
,
Parikh
P
,
von Pawel
J
,
Biesma
B
,
Vansteenkiste
J
,
Manegold
C
, et al
.
Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer
.
J Clin Oncol
2008
;
26
:
3543
51
.
36.
Reck
M
,
Rodriguez-Abreu
D
,
Robinson
AG
,
Hui
R
,
Csoszi
T
,
Fulop
A
, et al
.
Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer
.
N Engl J Med
2016
;
375
:
1823
33
.
37.
Douillard
JY
,
Siena
S
,
Cassidy
J
,
Tabernero
J
,
Burkes
R
,
Barugel
M
, et al
.
Final results from PRIME: randomized phase III study of panitumumab with FOLFOX4 for first-line treatment of metastatic colorectal cancer
.
Ann Oncol
2014
;
25
:
1346
55
.

Supplementary data