The CCTG PA.7 phase II trial of gemcitabine and nab-paclitaxel with or without durvalumab and tremelimumab as initial therapy in metastatic pancreatic ductal adenocarcinoma

Daniel J Renouf, Jonathan M Loree, Jennifer J Knox, James T Topham, Petr Kavan, Derek Jonker, Stephen Welch, Felix Couture, Frederic Lemay, Mustapha Tehfe, Mohammed Harb, Nathalie Aucoin, Yoo-Joung Ko, Patricia A Tang, Ravi Ramjeesingh, Brandon M Meyers, Christina A Kim, Pan Du, Shidong Jia, David F Schaeffer, Sharlene Gill, Dongsheng Tu, Chris J O'Callaghan, Daniel J Renouf, Jonathan M Loree, Jennifer J Knox, James T Topham, Petr Kavan, Derek Jonker, Stephen Welch, Felix Couture, Frederic Lemay, Mustapha Tehfe, Mohammed Harb, Nathalie Aucoin, Yoo-Joung Ko, Patricia A Tang, Ravi Ramjeesingh, Brandon M Meyers, Christina A Kim, Pan Du, Shidong Jia, David F Schaeffer, Sharlene Gill, Dongsheng Tu, Chris J O'Callaghan

Abstract

Immunotherapy-based monotherapy treatment in metastatic pancreatic ductal adenocarcinoma (mPDAC) has shown limited benefit outside of the mismatch repair deficiency setting, while safety and efficacy of combining dual-checkpoint inhibitor immunotherapy with chemotherapy remains uncertain. Here, we present results from the CCTG PA.7 study (NCT02879318), a randomized phase II trial comparing gemcitabine and nab-paclitaxel with and without immune checkpoint inhibitors durvalumab and tremelimumab in 180 patients with mPDAC. The primary endpoint was overall survival. Secondary endpoints included progression-free survival and objective response rate. Results of the trial were negative as combination immunotherapy did not improve survival among the unselected patient population (p = 0.72) and toxicity was limited to elevation of lymphocytes in the combination immunotherapy group (p = 0.02). Exploratory baseline circulating tumor DNA (ctDNA) sequencing revealed increased survival for patients with KRAS wildtype tumors in both the combination immunotherapy (p = 0.001) and chemotherapy (p = 0.004) groups. These data support the utility of ctDNA analysis in PDAC and the prognostic value of ctDNA-based KRAS mutation status.

Conflict of interest statement

P.D. and S.J. are employees of Predicine. The remaining authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Combination dual checkpoint inhibition and…
Fig. 1. Combination dual checkpoint inhibition and chemotherapy was not associated with OS, PFS nor ORR in an unselected population of patients with metastatic PDAC.
A Kaplan-Meier curve comparing overall survival (OS) between treatment arms. B Kaplan-Meier curve comparing progression-free survival (PFS) between treatment arms. C Forest plot showing results of subgroup analysis based on OS. Measure of center for error bars represents mean values. D Bar plot comparing differences in objective response rate (ORR) between treatment arms. Hazard ratio and confidence intervals (CIs) based on stratified Cox models are shown along with log-rank p values, and statistical tests were two-sided. Source data are provided as a source data file.
Fig. 2. Somatic mutation landscape of mPDAC…
Fig. 2. Somatic mutation landscape of mPDAC detected using liquid biopsy (ctDNA) sequencing at baseline.
A Oncoprint depicting most frequently mutated genes detected by baseline ctDNA sequencing in the cohort of 173 patients with mPDAC. Upper bars indicate blood tumor mutation burden (TMB; mut/Mb) levels and are colored based on microsatellite instability (red) or stable (grey) status. B Histogram with overlaid density curve showing distribution of blood tumor mutational burden levels across the cohort. C Histogram with overlaid density curve showing distribution of the number of SNV/indels detected by ctDNA sequencing. Source data are provided as a source data file.
Fig. 3. ctDNA-based KRAS mutation status is…
Fig. 3. ctDNA-based KRAS mutation status is highly prognostic in patients with mPDAC.
A Kaplan-Meier curve comparing overall survival (OS) between patients with KRAS wildtype (red) versus mutant (grey) tumors in the gemcitabine, nab-paclitaxel, durvalumab and tremelimumab (Gem+Nab-P+Durva+Treme) treatment group. B Kaplan-Meier curve comparing OS between patients with KRAS wildtype versus mutant tumors in the gemcitabine, nab-paclitaxel (Gem+Nab-P) treatment group. Hazard ratio and confidence intervals (CIs) based on stratified Cox models are shown along with log-rank p values, and statistical tests were two-sided. Source data are provided as a source data file.

References

    1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. Ca. Cancer J. Clin. 2021;71:7–33. doi: 10.3322/caac.21654.
    1. Conroy T, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med. 2011;364:1817–1825. doi: 10.1056/NEJMoa1011923.
    1. Von Hoff DD, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013;369:1691–1703. doi: 10.1056/NEJMoa1304369.
    1. O’Reilly EM, et al. Durvalumab with or without tremelimumab for patients with metastatic pancreatic ductal adenocarcinoma: a phase 2 randomized clinical trial. JAMA Oncol. 2019;5:1431–1438. doi: 10.1001/jamaoncol.2019.1588.
    1. Wainberg ZA, et al. Open-label, Phase I study of nivolumab combined with nab-paclitaxel plus gemcitabine in advanced pancreatic cancer. Clin. Cancer Res. J. Am. Assoc. Cancer Res. 2020;26:4814–4822. doi: 10.1158/1078-0432.CCR-20-0099.
    1. Marabelle A, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 Study. J. Clin. Oncol. J. Am. Soc. Clin. Oncol. 2020;38:1–10. doi: 10.1200/JCO.19.02105.
    1. Monteran L, Erez N. The dark side of fibroblasts: cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment. Front. Immunol. 2019;10:1835. doi: 10.3389/fimmu.2019.01835.
    1. Ford K, et al. NOX4 inhibition potentiates immunotherapy by overcoming cancer-associated fibroblast-mediated CD8 T-cell exclusion from tumors. Cancer Res. 2020;80:1846–1860. doi: 10.1158/0008-5472.CAN-19-3158.
    1. Alvarez R, et al. Stromal disrupting effects of nab-paclitaxel in pancreatic cancer. Br. J. Cancer. 2013;109:926–933. doi: 10.1038/bjc.2013.415.
    1. Miyashita T, et al. Neoadjuvant chemotherapy with gemcitabine plus nab-paclitaxel reduces the number of cancer-associated fibroblasts through depletion of pancreatic stroma. Anticancer Res. 2018;38:337–343.
    1. Antonia S, et al. Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study. Lancet Oncol. 2016;17:299–308. doi: 10.1016/S1470-2045(15)00544-6.
    1. Tsang ES, et al. Delving into early-onset pancreatic ductal adenocarcinoma: how does age fit in? Clin. Cancer Res. J. Am. Assoc. Cancer Res. 2021;27:246–254. doi: 10.1158/1078-0432.CCR-20-1042.
    1. Aung KL, et al. Genomics-driven precision medicine for advanced pancreatic cancer: early results from the COMPASS trial. Clin. Cancer Res. J. Am. Assoc. Cancer Res. 2018;24:1344–1354. doi: 10.1158/1078-0432.CCR-17-2994.
    1. Philip, P. A. et al. Molecular characterization of KRAS wild type tumors in patients with pancreatic adenocarcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. clincanres.3581.2021 (2022) 10.1158/1078-0432.CCR-21-3581.
    1. Jones MR, et al. NRG1 gene fusions are recurrent, clinically actionable gene rearrangements in KRAS wild-type pancreatic ductal adenocarcinoma. Clin. Cancer Res. J. Am. Assoc. Cancer Res. 2019;25:4674–4681. doi: 10.1158/1078-0432.CCR-19-0191.
    1. Heining C, et al. NRG1 fusions in KRAS wild-type pancreatic cancer. Cancer Discov. 2018;8:1087–1095. doi: 10.1158/-18-0036.
    1. Luchini C, et al. KRAS wild-type pancreatic ductal adenocarcinoma: molecular pathology and therapeutic opportunities. J. Exp. Clin. Cancer Res. CR. 2020;39:227. doi: 10.1186/s13046-020-01732-6.
    1. Herbreteau G, et al. Circulating tumor DNA as a prognostic determinant in small cell lung cancer patients receiving Atezolizumab. J. Clin. Med. 2020;9:E3861. doi: 10.3390/jcm9123861.
    1. Conroy JM, et al. Next generation sequencing of PD-L1 for predicting response to immune checkpoint inhibitors. J. Immunother. Cancer. 2019;7:18. doi: 10.1186/s40425-018-0489-5.
    1. Pender A, et al. Genome and transcriptome biomarkers of response to immune checkpoint inhibitors in advanced solid tumors. Clin. Cancer Res. J. Am. Assoc. Cancer Res. 2021;27:202–212. doi: 10.1158/1078-0432.CCR-20-1163.
    1. Tsujikawa T, et al. Quantitative multiplex immunohistochemistry reveals myeloid-inflamed tumor-immune complexity associated with poor prognosis. Cell Rep. 2017;19:203–217. doi: 10.1016/j.celrep.2017.03.037.
    1. Dougan SK. The pancreatic cancer microenvironment. Cancer J. Sudbury Mass. 2017;23:321–325. doi: 10.1097/PPO.0000000000000288.
    1. Zhu Y, et al. Diagnostic value of various liquid biopsy methods for pancreatic cancer: A systematic review and meta-analysis. Med. (Baltim.) 2020;99:e18581. doi: 10.1097/MD.0000000000018581.
    1. Laskin J, et al. NRG1 fusion-driven tumors: biology, detection, and the therapeutic role of afatinib and other ErbB-targeting agents. Ann. Oncol. J. Eur. Soc. Med. Oncol. 2020;31:1693–1703. doi: 10.1016/j.annonc.2020.08.2335.
    1. Windon AL, et al. A KRAS wild type mutational status confers a survival advantage in pancreatic ductal adenocarcinoma. J. Gastrointest. Oncol. 2018;9:1–10. doi: 10.21037/jgo.2017.10.14.
    1. Porta M, et al. Influence of KRAS mutations, persistent organic pollutants, and trace elements on survival from pancreatic ductal adenocarcinoma. Environ. Res. 2020;190:109781. doi: 10.1016/j.envres.2020.109781.
    1. Rachakonda PS, et al. Somatic mutations in exocrine pancreatic tumors: association with patient survival. PloS One. 2013;8:e60870. doi: 10.1371/journal.pone.0060870.
    1. McIntyre CA, et al. Alterations in driver genes are predictive of survival in patients with resected pancreatic ductal adenocarcinoma. Cancer. 2020;126:3939–3949. doi: 10.1002/cncr.33038.
    1. Wattenberg MM, et al. Platinum response characteristics of patients with pancreatic ductal adenocarcinoma and a germline BRCA1, BRCA2 or PALB2 mutation. Br. J. Cancer. 2020;122:333–339. doi: 10.1038/s41416-019-0582-7.
    1. O’Reilly EM, et al. Randomized, multicenter, phase ii trial of gemcitabine and cisplatin with or without veliparib in patients with pancreas adenocarcinoma and a germline BRCA/PALB2 Mutation. J. Clin. Oncol. J. Am. Soc. Clin. Oncol. 2020;38:1378–1388. doi: 10.1200/JCO.19.02931.
    1. Grant RC, et al. Clinical and genomic characterisation of mismatch repair deficient pancreatic adenocarcinoma. Gut. 2021;70:1894–1903. doi: 10.1136/gutjnl-2020-320730.
    1. O’Reilly EM, Hechtman JF. Tumour response to TRK inhibition in a patient with pancreatic adenocarcinoma harbouring an NTRK gene fusion. Ann. Oncol. J. Eur. Soc. Med. Oncol. 2019;30:viii36–viii40. doi: 10.1093/annonc/mdz385.
    1. Newman AM, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat. Biotechnol. 2016;34:547–555. doi: 10.1038/nbt.3520.
    1. Cibulskis K, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 2013;31:213–219. doi: 10.1038/nbt.2514.
    1. Gandara DR, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat. Med. 2018;24:1441–1448. doi: 10.1038/s41591-018-0134-3.
    1. Vandekerkhove G, et al. Circulating tumor DNA reveals clinically actionable somatic genome of metastatic bladder cancer. Clin. Cancer Res. J. Am. Assoc. Cancer Res. 2017;23:6487–6497. doi: 10.1158/1078-0432.CCR-17-1140.

Source: PubMed

3
Předplatit