Scheduling nab-paclitaxel combined with gemcitabine as first-line treatment for metastatic pancreatic adenocarcinoma

P G Corrie, W Qian, B Basu, J W Valle, S Falk, C Lwuji, H Wasan, D Palmer, M Scott-Brown, J Wadsley, S Arif, J Bridgewater, D Propper, R Gillmore, A Gopinathan, R Skells, P Bundi, R Brais, K Dalchau, L Bax, A Chhabra, A Machin, A Dayim, K McAdam, S Cummins, L Wall, R Ellis, A Anthoney, J Evans, Y T Ma, C Isherwood, A Neesse, D Tuveson, D I Jodrell, P G Corrie, W Qian, B Basu, J W Valle, S Falk, C Lwuji, H Wasan, D Palmer, M Scott-Brown, J Wadsley, S Arif, J Bridgewater, D Propper, R Gillmore, A Gopinathan, R Skells, P Bundi, R Brais, K Dalchau, L Bax, A Chhabra, A Machin, A Dayim, K McAdam, S Cummins, L Wall, R Ellis, A Anthoney, J Evans, Y T Ma, C Isherwood, A Neesse, D Tuveson, D I Jodrell

Abstract

Background: Nab-paclitaxel plus gemcitabine (nabP+gemcitabine) offers modest survival gains for patients with metastatic pancreatic ductal adenocarcinoma (PDAC). Sequential scheduling of nabP+gemcitabine in a PDAC mouse model improved efficacy; this hypothesis was tested in a clinical trial.

Methods: Patients with previously untreated metastatic PDAC were randomised to receive nabP+gemcitabine administered either concomitantly on the same day, or sequentially, with gemcitabine administered 24 h after nabP. The primary outcome measure was progression-free survival (PFS). Secondary outcome measures were objective response rate (ORR), overall survival (OS), safety, quality of life (QoL) and predictive biomarkers.

Results: In total, 71 patients received sequential (SEQ) and 75 concomitant (CON) treatment. Six-month PFS was 46% with SEQ and 32% with CON scheduling. Median PFS (5.6 versus 4.0 months, hazard ratio [HR] 0.67, 95% confidence interval [95% CI] 0.47-0.95, p = 0.022) and ORR (52% versus 31%, p = 0.023) favoured the SEQ arm; median OS was 10.2 versus 8.2 months (HR 0.93, 95% CI 0.65-1.33, p = 0.70). CTCAE Grade ≥3 neutropaenia incidence doubled with SEQ therapy but was not detrimental to QoL. Strongly positive tumour epithelial cytidine deaminase (CDA) expression favoured benefit from SEQ therapy (PFS HR 0.31, 95% CI 0.13-0.70).

Conclusions: SEQ delivery of nabP+gemcitabine improved PFS and ORR, with manageable toxicity, but did not significantly improve OS.

Clinical trial registration: ISRCTN71070888; ClinialTrials.gov (NCT03529175).

Conflict of interest statement

The following conflicts of interest are declared, which were incurred during the conduct of the SIEGE: Dr Corrie was the lead recipient of the SIEGE research grant from Celgene, which was paid to Cambridge University NHS Foundation Trust. Dr Basu and Dr Propper received funding from Celgene for research work outside of the SIEGE trial. Professor Bridgewater, Dr Corrie, Professor Evans, Professor Palmer, Professor Wadsley and Dr Wassan report personal fees from Celgene for advisory boards and/or speaker fees. Dr Falk, Professor Valle and Dr Basu received a travel grant from Celgene. Dr Propper was an expert witness when nab-Paclitaxel was submitted for NICE evaluation in 2017. Professor Evans is an editor of the clinical subject section of the British Journal of Cancer. Dr Falk and Dr Basu are Editorial Board members of the British Journal of Cancer. The remaining authors declare no competing interests.

Figures

Fig. 1. Kaplan-Meier curves of progression-free survival…
Fig. 1. Kaplan-Meier curves of progression-free survival (PFS) and overall survival (OS).
a, c PFS and (bd) OS for intention-to treat (ab) and evaluable (cd) patients.
Fig. 2. Changes from baseline in EORTC…
Fig. 2. Changes from baseline in EORTC QLQ-C30 QoL scores associated with treatment.
a Global health status (b) QoL subscales (c) symptoms.
Fig. 3. Box-plots demonstrating that baseline (pre-treatment)…
Fig. 3. Box-plots demonstrating that baseline (pre-treatment) cytidine deaminase (CDA) activity did not predict for objective response or toxicity experienced by individual patients during treatment.
a objective response (p of Kruskal–Wallis test = 0.62), (b) worst CTCAE grade experienced (p of Kruskal–Wallis test = 0.63) and (c) worst CTCAE neutropaenia grade experienced (p of Kruskal–Wallis test = 0.38).

References

    1. Malvezzi M, Bertuccio P, Levi F, La Vecchia C, Negri E. European cancer mortality predictions for the year 2014. Ann. Oncol. 2014;25:1650–1656. doi: 10.1093/annonc/mdu138.
    1. 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;369:1691–1703. doi: 10.1056/NEJMoa1304369.
    1. Goldstein D, El-Maraghi RH, Hammel P, Heinemann V, Kunzmann V, Sastre J, et al. Nab-Paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. J. Natl Cancer Inst. 2015;107:3–10.
    1. Von Hoff DD, Ramanathan RK, Borad MJ, Laheru DA, Smith LS, Wood TE, et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J. Clin. Oncol. 2011;29:4548–4554. doi: 10.1200/JCO.2011.36.5742.
    1. Alvarez R, Musteanu M, Garcia-garcia E, Lopez-Casas PP, Megias D, Guerra C, 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. Frese KK, Neesse A, Cook N, Bapiro TE, Lolkema MP, Jodrell DI, et al. Nab-Paclitaxel potentiates gemcitabine activity by reducing cytidine deaminase levels in a mouse model of pancreatic cancer. Cancer Discov. 2012;2:260–269. doi: 10.1158/-11-0242.
    1. Neesse A, Frese KK, Chan DS, Bapiro TE, Howat WJ, Richards FM, et al. SPARC independent drug delivery and antitumour effects of nab-paclitaxel in genetically engineered mice. Gut. 2014;63:974–983. doi: 10.1136/gutjnl-2013-305559.
    1. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457–1461. doi: 10.1126/science.1171362.
    1. Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013;62:112–120. doi: 10.1136/gutjnl-2012-302529.
    1. Ciccolini, J. Dosage de l’activite Serique en Cytidine Desaminase (CDA). PhD thesis, Universite d’Aix-Marseille, (2009).
    1. Osoba D, Rodrigues G, Myles J, Zee B, Pater J. Interpreting the significance of changes in health-related quality of life scores. J. Clin. Oncol. 1998;16:139–144. doi: 10.1200/JCO.1998.16.1.139.
    1. Reni M, Wan Y, Solem C, Whiting S, Ji X, Botteman M. Quality-adjusted survival with combination nab-paclitaxel+gemcitabine vs gemcitabine alone in metastatic pancreatic cancer: a Q-TWiST analysis. J. Med. Econ. 2014;17:338–346. doi: 10.3111/13696998.2014.903122.
    1. Chiritescu G, Dumon K, Verslype C, Houbiers G, Peeters M, Janssens J, et al. Final results of a phase II quality of life (QOL) randomized, cross-over (CO) study with gemcitabine (Gem) and nab-paclitaxel (n-P) in locally advanced and metastatic pancreatic cancer. Ann. Oncol. 2018;29(suppl_8):viii205–viii270.
    1. Serdjebi C, Seitz JF, Ciccolini J, Duluc M. Rapid deaminator status is associated with poor clinical outcome in pancreatic cancer patients treated with a gemcitabine-based regimen. Pharmacogenomics. 2013;14:1047–1051. doi: 10.2217/pgs.13.93.
    1. Carrato A, Falcone A, Ducreux M, Valle JW, Parnaby A, Djazouli K, et al. A systematic review of the burden of pancreatic cancer in Europe: Real-world impact on survival, quality of life and costs. J. Gastrointest. Cancer. 2015;46:201–211. doi: 10.1007/s12029-015-9724-1.
    1. Ciccolini J, Dahan L, Andre N, Evrard A, Duluc M, Yang AB, et al. Cytidine deaminase residual activity in serum is a predictive marker of early severe toxicities in adults after gemcitabine-based chemotherapies. J. Clin. Oncol. 2010;28:160–165. doi: 10.1200/JCO.2009.24.4491.
    1. Serdjebi C, Gagniere J, Desrame J, Fein F, Guimbaud R, Francois E, et al. FFCD-1004 clinical trial: impact of cytidine deaminase activity on clinical outcome in gemcitabine-monotherapy treated patients. PLoS ONE. 2015;10:e0135907. doi: 10.1371/journal.pone.0135907.
    1. Zauri M, Berridge G, Thezenas M-L, Pugh K, Goldin R, Kessler B, et al. CDA directs metabolism of epigenetic nucleosides revealing a therapeutic window in cancer. Nature. 2015;524:114–118. doi: 10.1038/nature14948.
    1. Tibaldi C, Giovannetti E, Vasile E, Mey V, Laan AC, Nannizzi S, et al. Correlation of CDA, ERCC1, and XPD polymorphisms with response and survival in gemcitabine/cisplatin-treated advanced non-small-cell lung cancer patients. Clin. Cancer Res. 2008;14:1797–1803. doi: 10.1158/1078-0432.CCR-07-1364.
    1. Yonemori K, Ueno H, Okusaka T, et al. Severe drug toxicity associated with a single-nucleotide polymorphism of the cytidine deaminase gene in a Japanese cancer patient treated with gemcitabine plus cisplatin. Clin. Cancer Res. 2005;11:2620–2624. doi: 10.1158/1078-0432.CCR-04-1497.
    1. Boyum A, Lovhaug D, Seeberg E, Nordlie EM. Identification of cytidine deaminase as an inhibitor of granulocyte-macrophage colony formation. Exp. Hematol. 1994;22:208–214.
    1. Micozzi D, Carpi FM, Pucciarelli S, Polzonetti V, Polidori P, Vilar S, et al. Human cytidine deaminase: a biochemical characterization of its naturally occurring variants. Int J. Biol. Macromol. 2014;63:64–74. doi: 10.1016/j.ijbiomac.2013.10.029.
    1. Hessmann E, Patzak MS, Klein L, Chen N, Kari V, Ramu I, et al. Fibroblast drug scavenging increases intratumoural gemcitabine accumulation in murine pancreas cancer. Gut. 2018;67:497–507. doi: 10.1136/gutjnl-2016-311954.
    1. Valkenburg KC, de Groot AE, Pienta KJ. Targeting the tumour stroma to improve cancer therapy. Nat. Rev. Clin. Oncol. 2018;15:366–381. doi: 10.1038/s41571-018-0007-1.
    1. Hidalgo M, Plaza C, Musteanu M, Illei P, Brachmann CB, Heise C, et al. SPARC expression did not predict efficacy of nab-paclitaxel plus gemcitabine or gemcitabine alone for metastatic pancreatic cancer in an exploratory analysis of the phase III MPACT trial. Clin. Cancer Res. 2015;21:4811–4818. doi: 10.1158/1078-0432.CCR-14-3222.

Source: PubMed

3
Prenumerera