Detection of Early-Stage Pancreatic Ductal Adenocarcinoma From Blood Samples: Results of a Multiplex Biomarker Signature Validation Study

Randall E Brand, Jan Persson, Svein Olav Bratlie, Daniel C Chung, Bryson W Katona, Alfredo Carrato, Marién Castillo, Julie Earl, Arto Kokkola, Aimee L Lucas, A James Moser, Corinne DeCicco, Linda Dexlin Mellby, Thomas C King, Randall E Brand, Jan Persson, Svein Olav Bratlie, Daniel C Chung, Bryson W Katona, Alfredo Carrato, Marién Castillo, Julie Earl, Arto Kokkola, Aimee L Lucas, A James Moser, Corinne DeCicco, Linda Dexlin Mellby, Thomas C King

Abstract

Introduction: The IMMray PanCan-d test combines an 8-plex biomarker signature with CA19-9 in a proprietary algorithm to detect pancreatic ductal adenocarcinoma (PDAC) in serum samples. This study aimed to validate the clinical performance of the IMMray PanCan-d test and to better understand test performance in Lewis-null (le/le) individuals who cannot express CA19-9.

Methods: Serum samples from 586 individuals were analyzed with the IMMray PanCan-d biomarker signature and CA19-9 assay, including 167 PDAC samples, 203 individuals at high risk of familial/hereditary PDAC, and 216 healthy controls. Samples were collected at 11 sites in the United States and Europe. The study was performed by Immunovia, Inc (Marlborough, MA), and sample identity was blinded throughout the study. Test results were automatically generated using validated custom software with a locked algorithm and predefined decision value cutoffs for sample classification.

Results: The IMMray PanCan-d test distinguished PDAC stages I and II (n = 56) vs high-risk individuals with 98% specificity and 85% sensitivity and distinguished PDAC stages I-IV vs high-risk individuals with 98% specificity and 87% sensitivity. We identified samples with a CA19-9 value of 2.5 U/mL or less as probable Lewis-null (le/le) individuals. Excluding these 55 samples from the analysis increased the IMMray PanCan-d test sensitivity to 92% for PDAC stages I-IV (n = 157) vs controls (n = 379) while maintaining specificity at 99%; test sensitivity for PDAC stages I and II increased from 85% to 89%.

Discussion: These results demonstrate the IMMray PanCan-d blood test can detect PDAC with high specificity (99%) and sensitivity (92%).

Trial registration: ClinicalTrials.gov NCT03693378.

Conflict of interest statement

Guarantor of the article: Thomas C. King, MD, PhD

Specific author contributions: R.E.B, J.P., S.O.B, D.C.C., B.W.K, A.C., M.C., J.E., A.K, A.L.L, A.J.M., and C.D.: collection of specimens and drafting of manuscript. L.D.M.: planning of study and review of manuscript. T.C.K: planning and conducting of study, interpreting of data, and drafting of manuscript. All authors have approved the final draft.

Financial support: The collection of samples for this study was supported by a grant from Immunovia AB through the PanFAM Clinical Trial (ClinicalTrials.gov Identifier: NCT03693378) to Drs. Brand, Katona, and Chung.

Potential competing interests: Drs. Lucas and Brand have also been compensated by Immunovia AB or Immunovia, Inc for their participation in scientific/medical meetings as outside speakers. Dr. Mellby is an employee of Immunovia AB, and Dr. King is an employee of Immunovia, Inc.

Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of The American College of Gastroenterology.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Distribution of decision values in the 3 cohorts.
Figure 2.
Figure 2.
ROC curve comparison between PDAC (early-stage and all-stage PDAC) and the healthy and PanFAM cohorts. PDAC, pancreatic ductal adenocarcinoma; ROC, receiver operating characteristic.
Figure 3.
Figure 3.
Decision values from Commercial Test Model Study.
Figure 4.
Figure 4.
ROC curve for IMMray PanCan-d test performance in PDAC vs all controls, excluding samples with CA19-9 values of 2.5 U/mL or less. PDAC, pancreatic ductal adenocarcinoma; ROC, receiver operating characteristic.
Figure 5.
Figure 5.
ROC curve for samples with CA19-9

References

    1. Kamisawa T, Wood LD, Itoi T, et al. Pancreatic cancer. Lancet 2016;388:73–85.
    1. Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. N Engl J Med 2014;371:1039–49.
    1. Rahib L, Smith BD, Aizenberg R, et al. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 2014;74(11):2913–21.
    1. Sohn TA, Yeo CJ, Cameron JL, et al. Resected adenocarcinoma of the pancreas-616 patients: Results, outcomes, and prognostic indicators. J Gastrointest Surg 2000;4(6):567–79.
    1. Canto MI, Kerdsirichairat T, Yeo CJ, et al. Surgical outcomes after pancreatic resection of screening-detected lesions in individuals at high-risk for developing pancreatic cancer. J Gastrointest Surg 2020;24(5):1101–10.
    1. Vasen H, Ibrahim I, Ponce CG, et al. Benefit of surveillance for pancreatic cancer in high-risk individuals: Outcome of long-term prospective followup studies from three European expert centers. J Clin Oncol 2016;34:2010–9.
    1. Canto MI, Almario JA, Schulick RD, et al. Risk of neoplastic progression in individuals at high risk for pancreatic cancer undergoing long-term surveillance. Gastroenterol 2018;155:740–51.
    1. Hanada K, Okazaki A, Hirano N, et al. Effective screening for early diagnosis of pancreatic cancer. Best Pract Res Clin Gastroenterol 2015;29:929–39.
    1. Chari ST, Kelly K, Hollingsworth MA, et al. Early detection of sporadic pancreatic cancer: Summative review. Pancreas 2015;44:693–712.
    1. Brentnall TA. Progress in the earlier detection of pancreatic cancer. J Clin Oncol 2016;34(17):1973–4.
    1. Everett JN, Burgos G, Chun J, et al. Cancer surveillance awareness and practice among families at increased risk for pancreatic adenocarcinoma. Cancer 2021;127:2271–8.
    1. Kawai S, Suzuki K, Nishio K, et al. Smoking and serum CA19-9 levels according to Lewis and secretor genotypes. Int J Cancer 2008;123:2880–4.
    1. Narimatsu H, Iwasaki H, Nakayama F, 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.
    1. Fahrmann JF, Schmidt CM, Mao X, et al. Lead-time trajectory of CA19-9 as an anchor marker for pancreatic cancer early detection. Gastroenterol 2021;160:1373–83.
    1. Roback JD Combs MR Grossman BJ et al.. In: Hillyer CD. (eds.). Technical Manual. 16th edn. AABB: Bethesda, MD, 2008.
    1. Okano K, Suzuki Y. Strategies for early detection of resectable pancreatic cancer. World J Gastroenterol 2014;20(32):11230–40.
    1. Locker GY, Hamilton S, Harris J, et al. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol 2006;24(33):5313–27.
    1. Galli C, Basso D, Plebani MCA. 19-9: Handle with care. Clin Chem Lab Med 2013;51(7):1369–83.
    1. Modi S, Kir D, Saluja AK. Old dog, new tricks: Use of CA 19-9 for early diagnosis of pancreatic cancer. Gastroenterol 2021;160:1019–21.
    1. Borrebaeck CAK. Precision diagnostics: Moving towards protein biomarker signatures of clinical utility in cancer. Nat Rev Cancer 2017;17:199–203.
    1. Hanash SM, Pitteri SJ, Faca VM. Mining the plasma proteome for cancer biomarkers. Nature 2008;452:571–9.
    1. Radon TP, Massat NJ, Jones R, et al. Identification of a three-biomarker panel in urine for early detection of pancreatic adenocarcinoma. Clin Cancer Res 2015;21(15):3512–21.
    1. Shaw VE, Lane B, Jenkinson C, et al. Serum cytokine biomarker panels for discriminating pancreatic cancer from benign pancreatic disease. Mol Cancer 2014;13:114.
    1. Mayers JR, Wu C, Clish CB, et al. Elevation of circulating branched chain amino acids is an early event in human pancreatic adenocarcinoma development. Nat Med 2014;20(10):1193–8.
    1. Jenkinson C, Elliott VL, Evans A, et al. Decreased serum thrombospondin-1 levels in pancreatic cancer patients up to 24 months prior to clinical diagnosis: Association with diabetes mellitus. Clin Cancer Res 2016;22(7):1734–43.
    1. Brand RE, Nolen BM, Zeh HJ, et al. Serum biomarker panels for the detection of pancreatic cancer. Clin Cancer Res 2011;17(4):805–16.
    1. Kim J, Barnlet WR, Oberg AL, et al. Detection of early pancreatic ductal adenocarcinoma with thrombospondin-2 and CA19-9 blood markers. Sci Transl Med 2017;9(398):eaah5583.
    1. Gerdtsson AS, Malats N, Säll A, et al. A multicenter trial defining a serum protein signature associated with pancreatic ductal adenocarcinoma. Int J Proteomics 2015:587250.
    1. Wingren C, Sandström A, Segersvärd R, et al. Identification of serum biomarker signatures associated with pancreatic cancer. Cancer Res 2012;72(10):2481–90.
    1. Mellby LD, Nyberg AP, Johansen JS, et al. Serum biomarker signature-based liquid biopsy for diagnosis of early-stage pancreatic cancer. J Clin Oncol 2018;36(28):2887–94.
    1. Amin MB, Edge S, Greene F, et al. . American Joint committee on cancer. AJCC Cancer Staging Manual. 8th edn. Springer: New York, NY, 2017.
    1. Delfani P, Carlsson A, King TC, et al. Commercial test model study – a multicenter study (2021). Immunovia AB website (). Accessed February 2021.
    1. Matsuno S, Egawa S, Fukuyama S, et al. Pancreatic cancer registry in Japan: 20 years of experience. Pancreas 2004;28:219–30.
    1. Shimizu Y, Yasui K, Matsueda K, et al. Small carcinoma of the pancreas is curable: New computed tomography finding, pathological study and postoperative results from a single institute. J Gastroenterol Hepatol 2005;20:1591–4.

Source: PubMed

Подписаться