Purpose:

CA19–9 synthesis is influenced by common variants in the fucosyltransferase (FUT) enzymes FUT3 and FUT2. We developed a clinical test to detect FUT variants, and evaluated its diagnostic performance for pancreatic ductal adenocarcinoma (PDAC).

Experimental Design:

A representative set of controls from the Cancer of the Pancreas Screening study was identified for each FUT functional group. Diagnostic sensitivity was determined first in a testing set of 234 PDAC cases, followed by a 134-case validation set, all of whom had undergone resection with curative intent without neoadjuvant therapy. Tumor marker gene testing was performed in the Johns Hopkins Molecular Diagnostics Laboratory. CA19–9 levels were measured in the Hopkins Clinical Chemistry lab. Receiver operating characteristic (ROC) curve analysis was used to evaluate the discriminative ability of CA19–9 alone versus with the gene test.

Results:

Applying the CA19–9 standard cutoff (<36 U/mL) to all 716 subjects yielded a 68.8% sensitivity in the test set of cases, 67.2% in the validation set, at 91.4% specificity. Applying 99th percentile cutoffs according to each individual's FUT group (3, 34.9, 41.8, and 89.2, for the FUT3-null, FUT-low, FUT-intermediate, and FUT-high groups, respectively) yielded a diagnostic sensitivity for CA19–9 in the first set of cases of 66.7%, 65.7% in the validation set, at 98.9% specificity. ROC analysis for CA19–9 alone yielded an AUC of 0.84; with the tumor marker gene test, AUC improved to 0.92 (P < 0.001).

Conclusions:

Using a tumor marker gene test to personalize an individual's CA19–9 reference range significantly improves diagnostic accuracy.

Translational Relevance

A highly accurate blood test could improve the early detection of pancreatic cancer. One step towards this goal is improving the diagnostic performance of CA19–9. Common inactivating variants in fucosyltransferase (FUT) genes involved in the CA19–9 synthetic pathway influence its levels in the circulation. These variants can be used to define an individual's functional FUT group, reflecting their predicted relative level of CA19–9 synthesis. We introduced a “CA19–9 tumor marker gene test” in the Johns Hopkins Molecular Diagnostics Laboratory as a companion test to serum CA19–9 and established reference ranges for each functional FUT group. Using a case/control design involving patients with resectable-stage pancreatic cancer and controls from the Cancer of the Pancreas Screening study, we find that using the tumor marker gene test to assign individuals their appropriate CA19–9 reference range significantly improved diagnostic performance.

Pancreatic cancer survival is slowly improving; 5-year survival is now ∼12% (1). Evidence from the Cancer of the Pancreas Screening (CAPS) study (2, 3) and other surveillance programs (4) indicates pancreatic imaging surveillance can improve early detection, especially the detection of Stage I pancreatic ductal adenocarcinoma (PDAC; ref. 2), and can reduce pancreatic cancer mortality. Nationally, the outcome of patients diagnosed and treated with Stage I PDAC is excellent (over 80% 5-year survival; ref. 5). However, some patients diagnosed with pancreatic cancer while under pancreatic surveillance will have higher-stage disease, and survival with PDAC depends heavily on the stage at diagnosis (6). The addition of blood-based biomarkers to pancreatic imaging surveillance could improve early detection. CA19–9 is the best available blood-based biomarker of pancreatic cancer and is used for monitoring disease status, but when studied as a diagnostic test achieves only ∼50% to 60% diagnostic sensitivity among patients with resectable-stage PDAC, at a specificity too low for diagnostic use (7).

The synthesis of CA19–9 and related metabolites involves multiple fucosyltransferases (FUT; ref. 8), and prior studies have reported that variants encoding the enzymes FUT2 and FUT3 influence CA19–9 levels (9–12). A functional FUT3 enzyme is needed to synthesize CA19–9, and subjects who lack functional FUT2 (known also as “non-secretors” for their lack of secretion of blood group antigens) have higher levels of CA19–9 than those with an intact FUT2 (12, 13). Control subjects who have only one functional FUT3 allele have lower levels of CA19–9 than those with two functional alleles; thus, individuals can be classified into one of four functional FUT groups that have different CA19–9 reference ranges (14). Assigning individuals their FUT group and CA19–9 reference range improved diagnostic performance (14) compared with the usual approach of applying the same standard cutoff for everyone.

We developed a “CA19–9 tumor marker gene test” in the Johns Hopkins Molecular Diagnostics Lab as a companion test to serum CA19–9 (see Supplementary Fig. S1). This tumor marker gene test was applied to control subjects enrolled in the CAPS study to establish clinical laboratory reference ranges for CA19–9 for each functional FUT group. We then applied this test to patients diagnosed with resectable-stage pancreatic cancer.

Subjects

This study included 716 subjects, including 348 high-risk individuals (HRI) and other controls enrolled in the Cancer of the Pancreas Screening-5 (CAPS5) study (NCT02000089) without evidence of pancreatic cancer at least 1 year after their blood-draw, and 368 patients diagnosed with PDAC that had blood samples collected prior to pancreatic resection at the Johns Hopkins Hospital, including a testing set of 234 cases from the Abe and colleagues study that had available biospecimen (14), and 134 Hopkins PDAC cases diagnosed between 2005 and 2019 that formed the validation set. Patients with PDAC who had undergone neoadjuvant chemotherapy were excluded. Controls were excluded if they had previous surgery for pancreatic cancer or high-grade dysplasia, or had worrisome imaging findings, or if they had other diseases or abnormalities known to be associated with an elevated CA19–9 [such as pancreatitis, an abnormally dilated main pancreatic duct or common bile duct, or large (>2 cm) or innumerable hepatic or renal cysts; ref. 7]. The CAPS controls selected for this study had undergone prior FUT genotyping in the Goggins lab and were selected to have sufficient numbers for each of the main FUT groups (approximately 100 subjects for each functional group, with fewer needed for the FUT3-null group as its reference range was expected to be close to 0). The final diagnosis of PDAC was made by surgical pathology using World Health Organization criteria (15). Tumor stage was defined by the American Joint Committee on Cancer (AJCC) 8th edition. All authors had access to study data and reviewed and approved the final manuscript. This study was performed in accordance with the Declaration of Helsinki, approved by the Johns Hopkins Institutional Review Board, and written informed consent was provided from all enrolled patients.

Tumor marker gene test

The tumor marker gene test was validated in the Johns Hopkins Molecular Diagnostics Laboratory. DNA samples were amplified using an Ampliseq custom primer pool panel sequenced on the Ion Torrent ion semiconductor sequencing platform similar to that used by Abe and colleagues (14). FUT variants were also sequenced using Sanger sequencing as an orthogonal method. Fifteen DNA samples were chosen from samples previously genotyped by Abe and colleagues (14) that contained one or more functionally impairing variants in either FUT2 or FUT3, including the common null variant in the FUT2 gene [rs601338, p.Trp154Ter, 461G > A (G428A in prior reference transcripts), (NM_000511.6)]; the weak FUT2 rs1047781 p.Ile140Phe, A418T variant (previously A385T) common in East Asians, which has five-fold less FUT2 enzyme activity (16); or one or more of seven FUT3 (NM_00149.3) variants (rs28362459, rs812936, rs778986, rs143012663, rs28362463, rs3745635, rs3894326), known to impair function alone, or in combination (17). One of these FUT3 null SNPs, (rs28362463; c.484G>A), was not in the Abe and colleagues study (14); it is rare in European populations, but relatively common in individuals with African heritage (∼12% minor allele frequency). Sanger sequencing was also used to characterize the FUT2 null variant, rs1047781. Prior studies have characterized multiple FUT3 haplotype groups (17), and phase analysis of 1000 Genomes data (18) identified 9 alleles including the wild-type (WT) allele. The population percentage of the variant haplotypes that affect function in the 1000 Genomes database for FUT3 and FUT2 is shown in Supplementary Table S1. An additional allele (FUT3508A) was found in our patient population but not called by phase analysis of 1000 Genomes. The FUT3 variant rs143012663 (c.445C > A) was not found in the 1000 Genomes data or in our study population. One allele (containing the rs28362459 minor allele without any other variants) has been described as hypofunctional (17). We classified this allele as functional. The possible FUT3 genotypes associated with these haplotypes are shown in Supplementary Table S2. Individuals were assigned to one of four functional groups based on the predicted function of each FUT3 allele and their FUT2 functional status: (i) FUT3 null (homozygous or compound heterozygous for nonfunctional FUT3 alleles); (ii) FUT3 heterozygous (one FUT3 null allele) with intact FUT2; (iii) FUT3 WT (no nonfunctional FUT3 variants), with intact FUT2; or (iv) FUT2 null, with intact FUT3 (individuals homozygous null or compound heterozygous for the weak/null FUT2 variants (rs601338 and/or rs1047781) with at least one FUT3 functional allele. For simplicity and based on the diagnostic cutoffs for each group, we renamed the groups, FUT3 null, FUT-low, FUT-intermediate and FUT-high. (In our prior paper, we referred to these groups as Group A, Group B-het, Group B-wild and Group C.)

All case and control DNA samples were sequenced and interpreted blinded to CA19–9 levels or to any prior genotype data. Libraries were sequenced using ion semiconductor sequencing technology on an Ion Torrent S5XL. Sequencing results were mapped to the reference human genome UCSC version hg19 (NCBI build GRCh37). BAM files were reviewed in IGV and genotypes interpreted and recalled using the IonTorrent Variant Caller software (ITVC, tvc5.10–10). Each individual was then assigned a FUT3 allelic group, and to one of the four FUT functional groups (Supplementary Fig. S1).

CA19–9 measurement

All serum samples were measured in the Johns Hopkins Clinical Chemistry Reference Laboratory using the Tosoh Bioscience ST-AIA-PACK CA 19–9 immunoassay method on the AIA 900 analyzer. The reference range of <36 U/mL was determined using healthy individuals.

Statistics

Summary statistics were used to describe the demographic, clinical and pathology information on all controls and PDAC cases. The PDAC cases were divided into a testing and validation set. Violin plots were used to present the distribution of CA19–9 in the controls based on the different FUT2/3 classes. The Fisher exact test and ANOVA were used to describe differences between FUT2/3 classes among the controls as appropriate. P values <0.05 were considered statistically significant. Estimated upper limits of the reference range for CA19–9 were calculated as 99th percentiles using nonparametric percentile of untransformed values using all data, including values <1 U/mL (considered as zero) with associated 90% confidence intervals (CI) calculated using the binomial distribution and a bootstrap approach. Receiver operating characteristic (ROC) curve analysis was used to evaluate discriminative ability of CA19–9 alone or CA19–9 with tumor marker gene test to distinguish PDAC cases from controls. Corresponding predictive probabilities were calculated from logistic regression models with CA19–9 as a continuous variable used alone or with FUT groups and terms for their interaction. GraphPad Prism version 9.5.0 for Windows (GraphPad Software, San Diego, California) and R version 4.2.1 were used for data analysis (RRID:SCR_001905).

Data sharing

The data generated in this study, including raw used to generate results, are available upon reasonable request from the corresponding author.

Tumor marker gene test development in the Johns Hopkins Molecular Diagnostics Laboratory

To validate the accuracy of the Johns Hopkins Molecular Diagnostics lab's CA19–9 tumor marker gene test at detecting variants in FUT2 and FUT3, a reference set of 15 DNA samples was analyzed that contained a representative set of FUT variants previously sequenced in the Goggins lab (14). Genotype interpretation was performed by J.R. Eshleman, who was blinded as to each sample's genotypes. The reproducibility of the assay was established by performing the next-generation sequencing (NGS) assay in triplicate on multiple days. The genotype calls were completely concordant across replicates, sequencing methods (all variants were confirmed by Sanger sequencing), and with the prior genotype calls on these samples by Abe and colleagues (14).

CA19–9 reference ranges in controls

There were 348 individuals who served as controls. Their characteristics are summarized in Table 1. There were no significant differences in the demographics between the FUT groups. DNA samples from controls were sequenced using the tumor marker genotype test in the Johns Hopkins Molecular Diagnostics lab and their genotypes interpreted independently by J.R. Eshleman, M. Dbouk, and Y. Ando, with the interpretation completely concordant. They were also completely concordant with prior sequencing results performed in the Goggins research lab.

Table 1.

Demographics and FUT2, FUT3 genotypes of CAPS control subjects.

CharacteristicTotal (N = 348)FUT3-null N = 49 (14.1%)FUT-low N = 105 (30.2%)FUT-intermediate N = 100 (28.7%)FUT-high N = 94 (27%)P valuea
Gender (Females); n (%) 182 (52.3%) 20 (40.8%) 51 (48.6%) 60 (60%) 51 (54.3%) 0.13 
Age; mean ±SD 59.5 ± 11 58.8 ± 10.7 58.2 ± 11.9 60.6 ± 11.4 60.2 ± 9.7 0.38 
Race/Ethnicity (white non-Hispanic vs. other) 311 (89.4%) 41 (83.7%) 89 (84.8%) 92 (92%) 89 (94.7%) 0.06 
Smoking  0.37 
  • Never

 
251 (72.1%) 36 (73.5%) 80 (76.2%) 74 (74%) 61 (64.9%)  
  • Former

 
86 (24.7%) 12 (24.5%) 22 (21%) 25 (25%) 27 (28.7%)  
  • Current

 
11 (3.2%) 1 (2%) 3 (2.9%) 1 (1%) 6 (6.4%)  
Pancreatic cyst(s) 118 (33.9%) 18 (36.7%) 37 (35.2%) 29 (29%) 34 (36.2%) 0.67 
CharacteristicTotal (N = 348)FUT3-null N = 49 (14.1%)FUT-low N = 105 (30.2%)FUT-intermediate N = 100 (28.7%)FUT-high N = 94 (27%)P valuea
Gender (Females); n (%) 182 (52.3%) 20 (40.8%) 51 (48.6%) 60 (60%) 51 (54.3%) 0.13 
Age; mean ±SD 59.5 ± 11 58.8 ± 10.7 58.2 ± 11.9 60.6 ± 11.4 60.2 ± 9.7 0.38 
Race/Ethnicity (white non-Hispanic vs. other) 311 (89.4%) 41 (83.7%) 89 (84.8%) 92 (92%) 89 (94.7%) 0.06 
Smoking  0.37 
  • Never

 
251 (72.1%) 36 (73.5%) 80 (76.2%) 74 (74%) 61 (64.9%)  
  • Former

 
86 (24.7%) 12 (24.5%) 22 (21%) 25 (25%) 27 (28.7%)  
  • Current

 
11 (3.2%) 1 (2%) 3 (2.9%) 1 (1%) 6 (6.4%)  
Pancreatic cyst(s) 118 (33.9%) 18 (36.7%) 37 (35.2%) 29 (29%) 34 (36.2%) 0.67 

aP values for the Fisher exact test for comparison of categorical measures and for ANOVA for comparison of age across genotypes.

The diagnostic cutoffs for the FUT3-null, FUT-low, FUT-intermediate and FUT-high groups for serum CA19–9 at the 99th percentile were <3, 34.9, 41.8, and 89.2 U/mL, respectively (see Table 2). Figure 1 shows violin plots of the distribution of CA19–9 levels for the overall control group as well as for each of the different FUT groups.

Table 2.

Serum CA19–9 reference ranges for FUT2/3 classes.

FUT2/3 functional groupFUT2/3 group definitionNMean ± SD (U/mL)99%-tile (U/mL)Nonparametric 90% CIBootstrapped 90% CI
FUT3-null FUT3-null 49 0.09 ± 0.47 <3a NA NA 
FUT-Low FUT2 WT/Het + FUT3 Het 105 11.63 ± 6.89 34.9 (30.3–35.7) (27.7–35.7) 
FUT-Intermediate FUT2 WT/Het + FUT3 WT 100 15.11 ± 7.92 41.8 (31.1–51.6) (30.7–51.6) 
FUT-High FUT2 null + FUT3 WT/Het 94 31.10 ± 14.28 89.2 (73.9–101.5) (67.7–101.5) 
FUT2/3 functional groupFUT2/3 group definitionNMean ± SD (U/mL)99%-tile (U/mL)Nonparametric 90% CIBootstrapped 90% CI
FUT3-null FUT3-null 49 0.09 ± 0.47 <3a NA NA 
FUT-Low FUT2 WT/Het + FUT3 Het 105 11.63 ± 6.89 34.9 (30.3–35.7) (27.7–35.7) 
FUT-Intermediate FUT2 WT/Het + FUT3 WT 100 15.11 ± 7.92 41.8 (31.1–51.6) (30.7–51.6) 
FUT-High FUT2 null + FUT3 WT/Het 94 31.10 ± 14.28 89.2 (73.9–101.5) (67.7–101.5) 

aActual cutoff was 2.8 U/mL; 47 subjects had undetectable CA19–9 (<1 U/mL), one had a level of 1.8, and another 2.8.

Figure 1.

Violin plots of the distribution of serum CA19–9 among controls for each FUT group. FUT groups are defined by inactivating variants in FUT2 and FUT3 genes responsible for the synthesis of CA19–9 and related molecules. All groups include all subjects without regard to their FUT group classification.

Figure 1.

Violin plots of the distribution of serum CA19–9 among controls for each FUT group. FUT groups are defined by inactivating variants in FUT2 and FUT3 genes responsible for the synthesis of CA19–9 and related molecules. All groups include all subjects without regard to their FUT group classification.

Close modal

We did not detect significant differences in the level of CA19–9 between other potential genotype subgroups (such as between FUT2 WT versus FUT2 functionally heterozygous controls that were FUT3 WT, or between FUT3 WT versus FUT3 heterozygous controls that were FUT2 null), either in our prior study (14) or in this study (Supplementary Table S3). There was also no significant difference in CA19–9 levels within each FUT group stratified by pancreatic cyst status (present/absent; data not shown). One third of the controls in this study had pancreatic cysts (Table 1). Pancreatic cysts in the controls were representative of what we find in patients who meet criteria for pancreas surveillance (19); almost all were subcentimeter in diameter, without concerning features (note as part of this study design, we excluded subjects with pancreatic cysts with worrisome imaging features).

Diagnostic performance of the CA19–9 tumor marker gene test in patients with PDAC versus controls

A summary of the characteristics of the PDAC cases is described in Table 3. The demographics of the controls were somewhat younger than the PDAC cases, reflecting the average age of patients who undergo surveillance (mean age 59.5 ± 11 years), with no significant difference between the groups with respect to gender or ethnicity (Supplementary Table S4). There was also no significant difference in the distribution of individual FUT variants in cases versus controls (Supplementary Table S5). The FUT2/FUT3 variant haplotypes of the cases and controls are listed in Supplementary Table S6.

Table 3.

Demographics, FUT2 and FUT3 genotypes, and tumor stage of the patients with PDAC.

Testing set (n = 234)Validation set (n = 134)
Gender (Females); n (%) 110 (47%) 59 (44%) 
Age; mean + SD 68 + 10.6 67.6 + 10.1 
Race/Ethnicity (white, non-Hispanic); n (%) 205 (87.6%) 121 (90.3%) 
Current or former smoker; n (%) 108 (46.2%) 65 (48.5%) 
FUT2 genotype; n (%)   
  • FUT2 WT

 
59 (25.2%) 35 (26.1%) 
  • FUT2 Heterozygous

 
118 (50.4%) 69 (51.5%) 
  • FUT2 Homozygous

 
57 (24.4%) 30 (22.4%) 
FUT3 genotype; n (%)   
  • FUT3 WT

 
100 (42.7%) 67 (50%) 
  • FUT3 Heterozygous

 
103 (44%) 56 (41.8%) 
  • FUT3 Null

 
31 (13.2%) 11 (8.2%) 
FUT2/3 Functional Group; n (%)   
  • FUT3-null

 
31 (13.2%) 11 (8.2%) 
  • FUT-Low

 
85 (36.3%) 46 (35.1%) 
  • FUT-Intermediate

 
68 (31.1%) 51 (38.1%) 
  • FUT-High

 
50 (21.4%) 26 (19.4%) 
PDAC Stage (AJCC 8th edition); n (%)   
  • Stage IA

 
19 (8.1%) 15 (11.2%) 
  • Stage IB

 
28 (12%) 16 (11.9%) 
  • Stage IIA

 
4 (1.7%) 4 (3%) 
  • Stage IIB

 
101 (43.2%) 47 (35.1%) 
  • Stage III

 
82 (35%) 52 (38.8%) 
Testing set (n = 234)Validation set (n = 134)
Gender (Females); n (%) 110 (47%) 59 (44%) 
Age; mean + SD 68 + 10.6 67.6 + 10.1 
Race/Ethnicity (white, non-Hispanic); n (%) 205 (87.6%) 121 (90.3%) 
Current or former smoker; n (%) 108 (46.2%) 65 (48.5%) 
FUT2 genotype; n (%)   
  • FUT2 WT

 
59 (25.2%) 35 (26.1%) 
  • FUT2 Heterozygous

 
118 (50.4%) 69 (51.5%) 
  • FUT2 Homozygous

 
57 (24.4%) 30 (22.4%) 
FUT3 genotype; n (%)   
  • FUT3 WT

 
100 (42.7%) 67 (50%) 
  • FUT3 Heterozygous

 
103 (44%) 56 (41.8%) 
  • FUT3 Null

 
31 (13.2%) 11 (8.2%) 
FUT2/3 Functional Group; n (%)   
  • FUT3-null

 
31 (13.2%) 11 (8.2%) 
  • FUT-Low

 
85 (36.3%) 46 (35.1%) 
  • FUT-Intermediate

 
68 (31.1%) 51 (38.1%) 
  • FUT-High

 
50 (21.4%) 26 (19.4%) 
PDAC Stage (AJCC 8th edition); n (%)   
  • Stage IA

 
19 (8.1%) 15 (11.2%) 
  • Stage IB

 
28 (12%) 16 (11.9%) 
  • Stage IIA

 
4 (1.7%) 4 (3%) 
  • Stage IIB

 
101 (43.2%) 47 (35.1%) 
  • Stage III

 
82 (35%) 52 (38.8%) 

We did not observe any significant difference in CA19–9 level according to age within any of the FUT groups (data not shown). Assigning each case and control to their appropriate CA19–9 reference range with the tumor marker gene test yielded a diagnostic sensitivity at 99th percentile cutoff of 66.7% (elevated in 157 of 234 PDAC cases in the testing set), with only 4 false-positives among the 348 controls, corresponding to a diagnostic specificity of 98.9% (Table 4; Supplementary Fig. S2). In contrast, using the uniform CA19–9 cutoff (<36 U/mL) for all the testing set PDAC cases yielded a sensitivity of 68%, but a specificity of 91.4%. Among Stage I cases diagnostic sensitivity using tumor marker gene test–determined cutoffs was 51.1% (Table 4). In the validation set of PDAC cases (Table 3), the diagnostic sensitivity of CA19–9 using tumor marker gene test–determined cutoffs was 65.7% (elevated in 88 of 134 cases), at 98.9% specificity (Table 4), compared with 67.2% sensitivity at 91.4% specificity using the uniform CA19–9 (36 U/mL) cutoff. The diagnostic sensitivity for Stage I PDAC cases in the validation set using the tumor marker gene test assigned reference range cutoffs was 41.9%.

Table 4.

Diagnostic performance of uniform versus stratified cutoffs for CA19–9.

Controls + testing set PDACsControls + validation set PDACs
All stages (I+II+III)Stage IAll stages (I+II+III)Stage I
UniformStratifiedUniformStratifiedUniformStratifiedUniformStratified
Sensitivity (95% CI) 68.8 (62.6–74.4) 66.7 (60.4–72.4) 53.2 (39.2–66.7) 51.1 (37.2–64.7) 67.2 (58.9–74.5) 65.7 (57.3–73.2) 51.6 (34.8–68.0) 41.9 (26.4–59.2) 
Specificity (95% CI) 91.4 (87.9–93.9) 98.9 (97.1–99.6) 91.4 (87.9–93.9) 98.9 (97.1–99.6) 91.4 (87.9–93.9) 98.9 (97.1–99.6) 91.4 (87.9–93.9) 98.9 (97.1–99.6) 
Controls + testing set PDACsControls + validation set PDACs
All stages (I+II+III)Stage IAll stages (I+II+III)Stage I
UniformStratifiedUniformStratifiedUniformStratifiedUniformStratified
Sensitivity (95% CI) 68.8 (62.6–74.4) 66.7 (60.4–72.4) 53.2 (39.2–66.7) 51.1 (37.2–64.7) 67.2 (58.9–74.5) 65.7 (57.3–73.2) 51.6 (34.8–68.0) 41.9 (26.4–59.2) 
Specificity (95% CI) 91.4 (87.9–93.9) 98.9 (97.1–99.6) 91.4 (87.9–93.9) 98.9 (97.1–99.6) 91.4 (87.9–93.9) 98.9 (97.1–99.6) 91.4 (87.9–93.9) 98.9 (97.1–99.6) 

Overall, the tumor marker gene test reclassified the CA19–9 result in 7.3% of the PDAC cases compared with CA19–9 alone (10 from negative-to-positive, 17 positive-to-negative). Among controls, 8.6% of subjects were reclassified, most reclassified from false-positive to true-negative (2 from negative-to-positive, 28 positive-to-negative; Supplementary Table S7).

Among all resectable-stage PDAC cases, the diagnostic sensitivity of the CA19–9 tumor marker gene test was 66.4%, and among Stage I PDAC cases, it was 46.4%.

ROC analysis of CA19–9 alone versus combined with tumor marker gene test

We also performed an ROC analysis to compare the ability of CA19–9 alone or with the tumor marker gene test to discriminate between PDAC cases and controls without using the above cutoffs (Fig. 2). For the combined cohort of the two sets of PDACs and the controls, the AUC for CA19–9 alone was 0.84 (95% CI, 0.81–0.87), while adding the tumor marker gene test to the model increased the AUC to 0.92 (95% CI, 0.90–0.94), a difference of 0.08 (95% CI, 0.05–0.11; P < 0.001). For Stage I PDAC cases, there was a similar AUC difference of 0.08 (95% CI, 0.04–0.13; P < 0.001); 0.77 (0.71–0.84) for CA19–9 alone versus 0.86 (0.81–0.91) when the tumor marker gene test is added to the model.

Figure 2.

ROC curve graphs and the AUC results for serum CA19–9 alone and with the tumor marker gene test (TMGT). The graph on the left includes the results for all patients with resectable-stage PDAC versus the CAPS study controls. On the right is the graph for patients with Stage I PDAC only.

Figure 2.

ROC curve graphs and the AUC results for serum CA19–9 alone and with the tumor marker gene test (TMGT). The graph on the left includes the results for all patients with resectable-stage PDAC versus the CAPS study controls. On the right is the graph for patients with Stage I PDAC only.

Close modal

We find that the diagnostic performance improves significantly when individuals are assigned the CA19–9 reference range that corresponds to their functional FUT group. The FUT group cutoffs include those both below and above the uniform CA19–9 cutoff (36 U/mL), so the FUT gene test identifies one of the main causes of both false-negative and false-positive CA19–9 tests. Assigning diagnostic cutoffs by FUT group yields much significantly higher diagnostic sensitivity at the same ∼99% specificity compared with the usual one size fits all approach to diagnostic cutoffs. Biomarkers are often assigned a uniformly high specificity cutoff, but the reduced diagnostic sensitivity that results arises particularly among patients with smaller, earlier-stage cancers that generate lower biomarker levels, the ones that when treated yield the most survival benefit (20, 21). On the other hand, the false-positives that result when the lower uniform cutoff is used for CA19–9 (our clinical lab cutoff is 36 U/mL) are mainly those that are FUT2-null; the upper limit of reference range for CA19–9 for FUT2-null subjects was 89.2 U/mL. Inevitably, although overall diagnostic performance was improved, the trade-offs between diagnostic sensitivity and specificity also resulted in more false-negatives in cases, mainly in the FUT-high group.

The FUT2 and FUT3 variants included in the CA19–9 tumor marker gene test are inactivating, common, and have large effects on CA19–9 levels. There is considerable interest in identifying and evaluating variants that influence expression of biomarkers and other proteins through genome-wide association studies (GWAS; refs. 10, 22) and other approaches. Many of the identified variants to date have only small biomarker effect sizes, though further study is needed. For pancreatic cancer, variants that associate with CA125, CEA, and CPA1 levels have been evaluated and have been shown to add to diagnostic accuracy, though their effects are more modest (14, 23).

In this study, we set diagnostic cutoffs to achieve 99% specificity so as to limit the number of false-positive tests. False-positives predominate when testing for a low prevalence disease like pancreatic cancer. The reference ranges for CA19–9 in this study were generated from subjects enrolled in the CAPS study, consisting mostly of patients undergoing pancreatic surveillance for their familial/genetic risk, who would be one of the main target populations of such a test. Even in the context of pancreatic surveillance, limiting false-positives is important as they create diagnostic confusion, worry, and unnecessary and potentially burdensome further testing. CA19–9 is not currently recommended as a stand-alone test for patients undergoing pancreatic surveillance, though it is often measured in patients with worrisome pancreatic imaging features as a possible indicator of invasive cancer (24, 25). CA19–9 has also been evaluated in initial studies as part of a multi-marker signature test (26). For the typical patient enrolled for pancreas surveillance such as CAPS with a relatively high lifetime pancreatic cancer risk (∼5% to 20%, depending on family history extent and germline mutation and other factors) the pretest probability of having pancreatic cancer detected at any given time is low. A typical CAPS patient might have a pretest probability of having a pancreas-imaging test-detected pancreatic cancer of only 0.25% (2). In this scenario, a patient with a positive CA19–9 with the tumor marker gene test (sensitivity 66%, specificity 99%) would have a negative predictive value of 99.8% and a positive predictive value for PDAC of ∼20%, with ∼4 of 5 positive CA19–9 tests being false-positive. This example highlights the importance of maintaining high diagnostic specificity cutoffs (∼99%) even in a high-risk cohort without any concerning findings. The consequences of false-positives tend to become more significant as downstream tests are performed, each with their own false-positive rate. It should be noted that the overall diagnostic yield of an elevated CA19–9 test in a CAPS cohort could be modestly higher than it is for PDAC alone as CA19–9 elevations also arise from other cancers (27), as well as from other diseases/abnormalities that can cause an elevated CA19–9 (28, 29).

With the CA19–9 tumor marker gene test available for clinical investigation, the next step is a study evaluating its diagnostic performance as an early detection test, and ultimately its diagnostic yield when added to existing pancreatic surveillance imaging tests. Patients enrolled into pancreas surveillance programs need long-term surveillance; for patients who might undergo regular CA19–9 tests over many years, adding a one-time tumor marker gene test would involve little additional cost. Until its diagnostic performance is better understood, the CA19–9 tumor marker gene test is best applied to patients who are undergoing surveillance pancreatic imaging, as abdominal imaging can identify many of the sources of benign elevations of CA19–9 (such as large hepatic, renal, or splenic cysts, biliary or pancreatic duct dilation associated with inflammation, etc.). Some of these diseases that cause elevated CA19–9 are uncommon in the CAPS surveillance setting, and they often cause symptoms (e.g., cholangitis, acute pancreatitis; ref. 30), and require diagnostic evaluation anyway. The clinical scenarios surrounding these diseases are very different to the scenario of an asymptomatic HRI undergoing tumor marker surveillance who presents with a positive tumor marker test, though it is important to evaluate the impact of false-positive tests in this setting in future studies

The goal of pancreatic surveillance is to detect early-stage pancreatic cancer, preferably when it is a small asymptomatic Stage I PDAC, or when there is high-grade dysplasia without invasive cancer (31), as intervening at these points in the natural history offers the best opportunities for cure. CA19–9 has also been evaluated in cohort study biorepositories (32–35), where evidence reveals elevations beginning mainly in the year prior to diagnosis, consistent with preclinical cancer progression. Perhaps the best criterion of a blood test for early detection is its diagnostic performance for Stage I PDAC. In our study, the CA19–9 tumor marker gene test had an overall diagnostic sensitivity for Stage I PDAC of 46.4%, at 98.9% specificity. Pancreatic imaging with EUS and/or MRI/CT detects many Stage I PDACs when the CA19–9 level is below even the tumor marker gene test defined cutoffs, so until more sensitive blood-based tests are available, pancreatic imaging will need to be the primary modality of pancreatic surveillance for CAPS subjects. How best to incorporate blood-based testing for patients undergoing pancreatic imaging surveillance remains to be determined, but one option would be to time the test in between annual pancreatic imaging tests.

There is allelic variation across different ancestral populations in the FUT3 and FUT2 null variants (rs601338 and rs1047781; refs. 36, 37). Genetic variation across populations is likely one of many factors influencing the diagnostic performance of CA19–9 when using a uniform cutoff. The tumor marker gene test can be expected to limit this source of variation. Some FUT2/FUT3 null variants are limited to certain population groups, though the overall prevalence of FUT3 and FUT2 null subjects in different populations does not vary nearly as much as individual variants. Common variants are often under evolutionary pressure, and there is evidence that FUT2 null alleles are under selection (38).Applying a FUT-based tumor marker gene test to other populations should take into account local variant prevalence.

There are limitations to this study. Our study was retrospective in nature. It allowed us to select similar numbers of controls from each of the functional FUT groups for the purposes of generating a reference range from our CAPS study population. As a result, the number of subjects in each functional group somewhat differs from the general population (for example, FUT2-null subjects represented a somewhat higher percentage of our controls, 27%, compared with the ∼20% to 22% prevalence in the U.S. population).

In conclusion, CA19–9 achieves greater diagnostic performance when combined with a tumor marker gene test that assigns individuals to the appropriate reference range for their FUT genotype group. The CA19–9 tumor marker genotype test merits prospective evaluation in HRIs undergoing pancreatic surveillance.

J. He reports grants from AbbVie outside the submitted work. L. Sokoll reports grants from NIH during the conduct of the study. No disclosures were reported by the other authors.

M. Dbouk: Formal analysis, investigation, visualization, writing–original draft, writing–review and editing. T. Abe: Investigation, visualization, writing–review and editing. C. Koi: Investigation, visualization, methodology, writing–review and editing. Y. Ando: Investigation, visualization, writing–review and editing. H. Saba: Data curation, writing–review and editing. E. Abou Diwan: Data curation, writing–review and editing. A. Macgregor-Das: Investigation, visualization, writing–review and editing. A.L. Blackford: Formal analysis, writing–review and editing. E. Mocci: Software, formal analysis, writing–review and editing. K. Beierl: Formal analysis, investigation, writing–review and editing. A. Dbouk: Data curation, writing–review and editing. J. He: Resources, writing–review and editing. R. Burkhart: Resources, writing–review and editing. A.M. Lennon: Resources, writing–review and editing. L. Sokoll: Investigation, writing–review and editing. M.I. Canto: Resources, supervision, writing–review and editing. J.R. Eshleman: Formal analysis, investigation, methodology, writing–review and editing. M. Goggins: Conceptualization, resources, supervision, funding acquisition, writing–original draft, project administration, writing–review and editing.

This work was supported by NIH grants (U01210170, R01CA176828 CA62924), Susan Wojcicki and Dennis Troper, and by a Stand Up To Cancer–Lustgarten Foundation Pancreatic Cancer Interception Translational Cancer Research Grant (Grant number: SU2C-AACR-DT25–17). The indicated SU2C research grant is administered by the American Association for Cancer Research, the scientific partner of SU2C.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Siegel
RL
,
Miller
KD
,
Wagle
NS
,
Jemal
A
.
Cancer statistics, 2023
.
CA Cancer J Clin
2023
;
73
:
17
48
.
2.
Dbouk
M
,
Katona
BW
,
Brand
RE
,
Chak
A
,
Syngal
S
,
Farrell
JJ
, et al
.
The multicenter cancer of pancreas screening study: impact on stage and survival
.
J Clin Oncol
2022
;
40
:
3257
66
.
3.
Canto
MI
,
Almario
JA
,
Schulick
RD
,
Yeo
CJ
,
Klein
A
,
Blackford
A
, et al
.
Risk of neoplastic progression in individuals at high risk for pancreatic cancer undergoing long-term surveillance
.
Gastroenterology
2018
;
155
:
740
51
.
4.
Klatte
DCF
,
Boekestijn
B
,
Wasser
MNJM
,
Feshtali Shahbazi
S
,
Ibrahim
IS
,
Mieog
JSD
, et al
.
Pancreatic cancer surveillance in carriers of a germline CDKN2A pathogenic variant: yield and outcomes of a 20-year prospective follow-up
.
J Clin Oncol
2022
;
40
:
3267
77
.
5.
Blackford
AL
,
Canto
MI
,
Klein
AP
,
Hruban
RH
,
Goggins
M
.
Recent trends in the incidence and survival of stage 1A pancreatic cancer: a surveillance, epidemiology, and end results analysis
.
J Natl Cancer Inst
2020
;
112
:
1162
9
.
6.
Overbeek
KA
,
Levink
IJM
,
Koopmann
BDM
,
Harinck
F
,
Konings
ICAW
,
Ausems
MGEM
, et al
.
Long-term yield of pancreatic cancer surveillance in high-risk individuals
.
Gut
2021
;
5
:
2020
323611
.
7.
Galli
C
,
Basso
D
,
Plebani
M
.
CA 19–9: handle with care
.
Clin Chem Lab Med
2013
;
51
:
1369
83
.
8.
Engle
DD
,
Tiriac
H
,
Rivera
KD
,
Pommier
A
,
Whalen
S
,
Oni
TE
, et al
.
The glycan CA19–9 promotes pancreatitis and pancreatic cancer in mice
.
Science
2019
;
364
:
1156
62
.
9.
Luo
G
,
Guo
M
,
Jin
K
,
Liu
Z
,
Liu
C
,
Cheng
H
, et al
.
Optimize CA19–9 in detecting pancreatic cancer by Lewis and secretor genotyping
.
Pancreatology
2016
;
16
:
1057
62
.
10.
He
M
,
Wu
C
,
Xu
J
,
Guo
H
,
Yang
H
,
Zhang
X
, et al
.
A genome wide association study of genetic loci that influence tumor biomarkers cancer antigen 19–9, carcinoembryonic antigen and α fetoprotein and their associations with cancer risk
.
Gut
2014
;
63
:
143
51
.
11.
Narimatsu
H
,
Iwasaki
H
,
Nakayama
F
,
Ikehara
Y
,
Kudo
T
,
Nishihara
S
, et al
.
Lewis and secretor gene dosages affect CA19–9 and DU-PAN-2 serum levels in normal individuals and colorectal cancer patients
.
Cancer Res
1998
;
58
:
512
8
.
12.
Kawai
S
,
Suzuki
K
,
Nishio
K
,
Ishida
Y
,
Okada
R
,
Goto
Y
, et al
.
Smoking and serum CA19–9 levels according to Lewis and secretor genotypes
.
Int J Cancer
2008
;
123
:
2880
4
.
13.
Vestergaard
EM
,
Hein
HO
,
Meyer
H
,
Grunnet
N
,
Jørgensen
J
,
Wolf
H
, et al
.
Reference values and biological variation for tumor marker CA 19–9 in serum for different Lewis and secretor genotypes and evaluation of secretor and Lewis genotyping in a Caucasian population
.
Clin Chem
1999
;
45
:
54
61
.
14.
Abe
T
,
Koi
C
,
Kohi
S
,
Song
K-B
,
Tamura
K
,
Macgregor-Das
A
, et al
.
Gene variants that affect levels of circulating tumor markers increase identification of patients with pancreatic cancer
.
Clin Gastroenterol Hepatol
2020
;
18
:
1161
9
.
15.
Bosman
FT
,
Carneiro
F
,
Hruban
RH
,
Theise
ND
.
editors
WHO classification of tumours of the digestive system
. 4th ed.
Lyon, France
;
2010
.
16.
Henry
S
,
Mollicone
R
,
Fernandez
P
,
Samuelsson
B
,
Oriol
R
,
Larson
G
, et al
.
Molecular basis for erythrocyte Le(a+ b+) and salivary ABH partial-secretor phenotypes: expression of a FUT2 secretor allele with an A–&gt;T mutation at nucleotide 385 correlates with reduced alpha(1,2) fucosyltransferase activity
.
Glycoconj J
1996
;
13
:
985
93
.
17.
Ørntoft
TF
,
Vestergaard
EM
,
Holmes
E
,
Jakobsen
JS
,
Grunnet
N
,
Mortensen
M
, et al
.
Influence of Lewis alpha1–3/4-L-fucosyltransferase (FUT3) gene mutations on enzyme activity, erythrocyte phenotyping, and circulating tumor marker sialyl-Lewis a levels
.
J Biol Chem
1996
;
271
:
32260
8
.
18.
Mocci
E
,
Debeljak
M
,
Klein
AP
,
Eshleman
JR
.
A new fast phasing method based on haplotype subtraction
.
J Mol Diagn
2019
;
21
:
427
36
.
19.
Canto
MI
,
Hruban
RH
,
Fishman
EK
,
Kamel
IR
,
Schulick
R
,
Zhang
Z
, et al
.
Frequent detection of pancreatic lesions in asymptomatic high-risk individuals
.
Gastroenterology
2012
;
142
:
796
804
;
quiz e14–5
.
20.
Cohen
JD
,
Javed
AA
,
Thoburn
C
,
Wong
F
,
Tie
J
,
Gibbs
P
, et al
.
Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers
.
Proc Natl Acad Sci USA
2017
;
114
:
10202
7
.
21.
Kim
J
,
Bamlet
WR
,
Oberg
AL
,
Chaffee
KG
,
Donahue
G
,
Cao
X-J
, et al
.
Detection of early pancreatic ductal adenocarcinoma with thrombospondin-2 and CA19–9 blood markers
.
Sci Transl Med
2017
;
9
:
eaah5583
.
22.
Olafsson
S
,
Alexandersson
KF
,
Gizurarson
JGK
,
Hauksdottir
K
,
Gunnarsson
O
,
Olafsson
K
, et al
.
Common and rare sequence variants influencing tumor biomarkers in blood
.
Cancer Epidemiol Biomarkers Prev
2019
;
29
:
225
35
.
23.
Tanaka
H
,
Tamura
K
,
Abe
T
,
Yoshida
T
,
Macgregor-Das
A
,
Dbouk
M
, et al
.
Serum carboxypeptidase activity and genotype-stratified CA19–9 to detect early-stage pancreatic cancer
.
Clin Gastroenterol Hepatol
2021
;
12
:
01094
6
.
24.
Ciprani
D
,
Morales-Oyarvide
V
,
Qadan
M
,
Hank
T
,
Weniger
M
,
Harrison
JM
, et al
.
An elevated CA 19–9 is associated with invasive cancer and worse survival in IPMN
.
Pancreatology
2020
;
20
:
729
35
.
25.
Hirono
S
,
Kawai
M
,
Okada
K-I
,
Miyazawa
M
,
Shimizu
A
,
Kitahata
Y
, et al
.
Factors associated with invasive intraductal papillary mucinous carcinoma of the pancreas
.
JAMA Surg
2017
;
152
:
e165054
.
26.
Brand
RE
,
Persson
J
,
Bratlie
SO
,
Chung
DC
,
Katona
BW
,
Carrato
A
, et al
.
Detection of early-stage pancreatic ductal adenocarcinoma from blood samples: results of a multiplex biomarker signature validation study
.
Clin Transl Gastroenterol
2022
;
13
:
e00468
.
27.
Wannhoff
A
,
Hov
JR
,
Folseraas
T
,
Rupp
C
,
Friedrich
K
,
Anmarkrud
JA
, et al
.
FUT2 and FUT3 genotype determines CA19–9 cut-off values for detection of cholangiocarcinoma in patients with primary sclerosing cholangitis
.
J Hepatol
2013
;
59
:
1278
84
.
28.
Passerini
R
,
Cassatella
MC
,
Boveri
S
,
Salvatici
M
,
Radice
D
,
Zorzino
L
, et al
.
The pitfalls of CA19–9: routine testing and comparison of two automated immunoassays in a reference oncology center
.
Am J Clin Pathol
2012
;
138
:
281
7
.
29.
Kim
S
,
Park
BK
,
Seo
JH
,
Choi
J
,
Choi
JW
,
Lee
CK
, et al
.
Carbohydrate antigen 19–9 elevation without evidence of malignant or pancreatobiliary diseases
.
Sci Rep
2020
;
10
:
8820
.
30.
Brand
RE
,
Nolen
BM
,
Zeh
HJ
,
Allen
PJ
,
Eloubeidi
MA
,
Goldberg
M
, et al
.
Serum biomarker panels for the detection of pancreatic cancer
.
Clin Cancer Res
2011
;
17
:
805
16
.
31.
Goggins
M
,
Overbeek
KA
,
Brand
R
,
Syngal
S
,
Del Chiaro
M
,
Bartsch
DK
, et al
.
Management of patients with increased risk for familial pancreatic cancer: updated recommendations from the international Cancer of the Pancreas Screening (CAPS) Consortium
.
Gut
2020
;
69
:
7
17
.
32.
Fahrmann
JF
,
Schmidt
CM
,
Mao
X
,
Irajizad
E
,
Loftus
M
,
Zhang
J
, et al
.
Lead-time trajectory of CA19–9 as an anchor marker for pancreatic cancer early detection
.
Gastroenterology
2021
;
160
:
1373
83
.
33.
O'Brien
DP
,
Sandanayake
NS
,
Jenkinson
C
,
Gentry-Maharaj
A
,
Apostolidou
S
,
Fourkala
E-O
, et al
.
Serum CA19–9 is significantly upregulated up to 2 years before diagnosis with pancreatic cancer: implications for early disease detection
.
Clin Cancer Res
2015
;
21
:
622
31
.
34.
Honda
K
,
Katzke
VA
,
Hüsing
A
,
Okaya
S
,
Shoji
H
,
Onidani
K
, et al
.
CA19–9 and apolipoprotein-A2 isoforms as detection markers for pancreatic cancer: a prospective evaluation
.
Int J Cancer
2019
;
144
:
1877
87
.
35.
Nolen
BM
,
Brand
RE
,
Prosser
D
,
Velikokhatnaya
L
,
Allen
PJ
,
Zeh
HJ
, et al
.
Prediagnostic serum biomarkers as early detection tools for pancreatic cancer in a large prospective cohort study
.
PLoS One
2014
;
9
:
e94928
.
36.
Cakir
B
,
Pankow
JS
,
Salomaa
V
,
Couper
D
,
Morris
TL
,
Brantley
KR
, et al
.
Distribution of Lewis (FUT3)genotype and allele: frequencies in a biethnic United States population
.
Ann Hematol
2002
;
81
:
558
65
.
37.
Soejima
M
,
Koda
Y
.
Survey and characterization of nonfunctional alleles of FUT2 in a database
.
Sci Rep
2021
;
11
:
3186
.
38.
Ferrer-Admetlla
A
,
Sikora
M
,
Laayouni
H
,
Esteve
A
,
Roubinet
F
,
Blancher
A
, et al
.
A natural history of FUT2 polymorphism in humans
.
Mol Biol Evol
2009
;
26
:
1993
2003
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.