Safety and activity of the TGFβ receptor I kinase inhibitor galunisertib plus the anti-PD-L1 antibody durvalumab in metastatic pancreatic cancer

Davide Melisi, Do-Youn Oh, Antoine Hollebecque, Emiliano Calvo, Anna Varghese, Erkut Borazanci, Teresa Macarulla, Valeria Merz, Camilla Zecchetto, Yumin Zhao, Ivelina Gueorguieva, Michael Man, Leena Gandhi, Shawn T Estrem, Karim A Benhadji, Mark C Lanasa, Emin Avsar, Susan C Guba, Rocio Garcia-Carbonero, Davide Melisi, Do-Youn Oh, Antoine Hollebecque, Emiliano Calvo, Anna Varghese, Erkut Borazanci, Teresa Macarulla, Valeria Merz, Camilla Zecchetto, Yumin Zhao, Ivelina Gueorguieva, Michael Man, Leena Gandhi, Shawn T Estrem, Karim A Benhadji, Mark C Lanasa, Emin Avsar, Susan C Guba, Rocio Garcia-Carbonero

Abstract

Background: We assessed the safety, efficacy, and pharmacokinetics of the transforming growth factor beta (TGFβ) receptor inhibitor galunisertib co-administered with the anti-programmed death-ligand 1 (PD-L1) antibody durvalumab in recurrent/refractory metastatic pancreatic cancer previously treated with ≤2 systemic regimens.

Methods: This was a two-part, single-arm, multinational, phase Ib study. In a dose-finding phase, escalating oral doses of galunisertib were co-administered on days 1-14 with fixed-dose intravenous durvalumab 1500 mg on day 1 every 4 weeks (Q4W), followed by an expansion cohort phase.

Results: The galunisertib recommended phase II dose (RP2D) when co-administered with durvalumab 1500 mg Q4W was 150 mg two times per day. No dose-limiting toxicities were recorded. Among 32 patients treated with galunisertib RP2D, 1 patient had partial response, 7 had stable disease, 15 had objective progressive disease, and 9 were not evaluable. Disease control rate was 25.0%. Median overall survival and progression-free survival were 5.72 months (95% CI: 4.01 to 8.38) and 1.87 months (95% CI: 1.58 to 3.09), respectively. Pharmacokinetic profiles for combination therapy were comparable to those published for each drug. There was no association between potential biomarkers and treatment outcomes.

Conclusion: Galunisertib 150 mg two times per day co-administered with durvalumab 1500 mg Q4W was tolerable. Clinical activity was limited. Studying this combination in patients in an earlier line of treatment or selected for predictive biomarkers of TGFβ inhibition might be a more suitable approach.

Trial registration number: ClinicalTrials.gov identifier: NCT02734160.

Keywords: clinical trials as topic; immunotherapy; investigational; therapies; tumor biomarkers; tumor microenvironment.

Conflict of interest statement

Competing interests: DM has received research funding from Celgene, Incyte, and Shire, and has a consulting role with Baxter, Eli Lilly and Company, Incyte, and Shire. D-YO has received research funding from Array, AstraZeneca, Eli Lilly and Company, and Novartis, and has a consulting/advisory role with ASLAN, AstraZeneca, Bayer, Celgene, Genentech/Roche, Halozyme, Merck Serono, Novartis, Taiho, and Zymeworks. AH has received travel and accommodation expenses from Eli Lilly and Company. EC has received research funding from AbbVie, Amcure, Amgen, AstraZeneca, BeiGene, BMS, Boehringer-Ingelheim, CytomX, Eli Lilly and Company, H3, Incyte, Kura, LOXO, Macrogenics, Menarini, Merck, Merck Serono, Merus, Millennium Pharmaceuticals, Nanobiotix Janssen, Nektar, Novartis, Pfizer, PharmaMar, Principia Bayer, PsiOxus Therapeutics, PUMA, Rigontec, Roche/Genentech, Sanofi, Tahio, Tesaro; has a consulting/advisory role with AbbVie, Amcure, AstraZeneca, Boehringer-Ingelheim, Celgene, Cerulean Pharma, EUSA, GLG, Guidepoint Global, Janssen-Cilag, Nanobiotix Janssen, Novartis, Pfizer, Pierre Pharma, PsiOxus Therapeutics, Roche/Genentech, Seattle Genetics, Servier; is employed by HM Hospitals Group and START; has shares in HM Hospitals Group, International Cancer Consultants, Oncoart Associated, and START; and is president and founder of NPO Foundation Intheos (Investigational Therapeutics in Oncological Sciences). AMV has participated in clinical trials funded by Bristol Myers Squibb, Eli Lilly and Company, GlaxoSmithKline, Silenseed, and Verastem. EB has received honoraria for consultancy from Corcept Therapeutics and Invitae, is on the speaker bureau for Ipsen, and his institution has received research funding from Biontech, Bristol-Myers Squibb, Daiichi Sankyo, Eli Lilly and Company, Helix, Mabvax, Merck, Minneamrita Therapeutics, Pharmacyclics, and Samumed. TM has received honoraria for consultancy from Baxalta, Baxter, Celgene, Genzyme, Roche, Sanofi, Shire Pharmaceuticals, Tesaro, and QED Therapeutics, and has received travel/accommodation compensation from Bayer, H3 Biomedicine, Merck, and Sanofi. VM and CZ have no conflicts of interest to disclose. YZ, IG, MM, LG, STE, and EA are employees and stock holders of Eli Lilly and Company. KAB was an employee of Eli Lilly and Company at the time this research was conducted and is a stock holder of Eli Lilly and Company and a current employee of Taiho Oncology. MCL is a current employee and stock holder of AstraZeneca. SCG was an employee of Eli Lilly and Company at the time this research was conducted and is a stock holder of Eli Lilly and Company. RG-C declares having provided scientific advice and/or received honoraria from AAA, Advanz Pharma, Amgen, Bayer, BMS, Eli Lilly and Company, HMP, Ipsen, Merck, Midatech Pharma, MSD, Novartis, PharmaMar, Pfizer, Roche, and Sanofi, and has received research support from Pfizer and BMS. Work in the unit of DM was partially supported by the Associazione Italiana per la Ricerca sul Cancro (AIRC) Investigator Grant n°23719 and 5x1000 Grant n°12182, by the Italian Ministry of Health Ricerca Finalizzata 2016 GR-2016- 02361134 grant, and by the patients associations 'Nastro Viola' and 'Voglio il Massimo' donations.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Best response by patient based on change in tumor size. All treated patients with best overall response (n=33). Blue number=percentage of tumor cells at baseline with positive PD-L1 membrane staining. Dashed lines at –30% and 20% represent the thresholds for SD; neither an increase in size of more than 20% nor a decrease in size of more than 30% since the initial baseline measurement. BID, two times per day; PD, progressive disease; PD-L1, programmed death-ligand 1; PR, partial response; QD, one time per day; SD, stable disease.
Figure 2
Figure 2
Association between PD-L1 expression and clinical benefit. For the purpose of the biomarker analysis, any patient who had a CR, PR, or SD for ≥3 months was considered as having derived clinical benefit from study treatment. CR, complete response; NE, non-evaluable; PD, progressive disease; PD-L1, programmed death-ligand 1; PR, partial response; SD, stable disease.
Figure 3
Figure 3
(A) Overall survival and (B) progression-free survival in the galunisertib 150 mg BID + durvalumab 1500 mg Q4W group. BID, two times a day; Q4W, every 4 weeks.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020;70:7–30. 10.3322/caac.21590
    1. Rawla P, Sunkara T, Gaduputi V. Epidemiology of pancreatic cancer: global trends, etiology and risk factors. World J Oncol 2019;10:10–27. 10.14740/wjon1166
    1. Macdonald S, Mair F. Tackling cancers of unmet need: the pancreatic cancer pathway. Lancet Gastroenterol Hepatol 2016;1:266–7. 10.1016/S2468-1253(16)30113-3
    1. Hilmi M, Bartholin L, Neuzillet C. Immune therapies in pancreatic ductal adenocarcinoma: where are we now? World J Gastroenterol 2018;24:2137–51. 10.3748/wjg.v24.i20.2137
    1. Oberstein PE, Olive KP. Pancreatic cancer: why is it so hard to treat? Therap Adv Gastroenterol 2013;6:321–37. 10.1177/1756283X13478680
    1. Bailey P, Chang DK, Nones K, et al. . Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016;531:47–52. 10.1038/nature16965
    1. Massagué J. TGFβ signalling in context. Nat Rev Mol Cell Biol 2012;13:616–30. 10.1038/nrm3434
    1. Pickup M, Novitskiy S, Moses HL. The roles of TGFβ in the tumour microenvironment. Nat Rev Cancer 2013;13:788–99. 10.1038/nrc3603
    1. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 2015;27:450–61. 10.1016/j.ccell.2015.03.001
    1. Henriksen A, Dyhl-Polk A, Chen I, et al. . Checkpoint inhibitors in pancreatic cancer. Cancer Treat Rev 2019;78:17–30. 10.1016/j.ctrv.2019.06.005
    1. Royal RE, Levy C, Turner K, et al. . Phase 2 trial of single agent ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother 2010;33:828–33. 10.1097/CJI.0b013e3181eec14c
    1. Eso Y, Shimizu T, Takeda H, et al. . Microsatellite instability and immune checkpoint inhibitors: toward precision medicine against gastrointestinal and hepatobiliary cancers. J Gastroenterol 2020;55:15–26. 10.1007/s00535-019-01620-7
    1. Lee V, Murphy A, Le DT, et al. . Mismatch repair deficiency and response to immune checkpoint blockade. Oncologist 2016;21:1200–11. 10.1634/theoncologist.2016-0046
    1. Prasad V, Kaestner V, Mailankody S. Cancer drugs approved based on biomarkers and not tumor type-FDA approval of pembrolizumab for mismatch repair-deficient solid cancers. JAMA Oncol 2018;4:157–8. 10.1001/jamaoncol.2017.4182
    1. Bazzichetto C, Conciatori F, Luchini C, et al. . From genetic alterations to tumor microenvironment: the Ariadne’s string in pancreatic cancer. Cells 2020;9:309. 10.3390/cells9020309
    1. Sideras K, Braat H, Kwekkeboom J, et al. . Role of the immune system in pancreatic cancer progression and immune modulating treatment strategies. Cancer Treat Rev 2014;40:513–22. 10.1016/j.ctrv.2013.11.005
    1. Martinez-Bosch N, Vinaixa J, Navarro P. Immune evasion in pancreatic cancer: from mechanisms to therapy. Cancers (Basel) 2018;10:6. 10.3390/cancers10010006
    1. Principe DR, DeCant B, Mascariñas E, et al. . TGFβ signaling in the pancreatic tumor microenvironment promotes fibrosis and immune evasion to facilitate tumorigenesis. Cancer Res 2016;76:2525–39. 10.1158/0008-5472.CAN-15-1293
    1. Kabacaoglu D, Ciecielski KJ, Ruess DA, et al. . Immune checkpoint inhibition for pancreatic ductal adenocarcinoma: current limitations and future options. Front Immunol 2018;9:1878. 10.3389/fimmu.2018.01878
    1. Kowal J, Kornete M, Joyce JA. Re-education of macrophages as a therapeutic strategy in cancer. Immunotherapy 2019;11:677–89. 10.2217/imt-2018-0156
    1. Herbertz S, Sawyer JS, Stauber AJ, et al. . Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther 2015;9:4479–99. 10.2147/DDDT.S86621
    1. Melisi D, Garcia-Carbonero R, Macarulla T, et al. . Galunisertib plus gemcitabine vs. gemcitabine for first-line treatment of patients with unresectable pancreatic cancer. Br J Cancer 2018;119:1208–14. 10.1038/s41416-018-0246-z
    1. Gueorguieva I, Tabernero J, Melisi D, et al. . Population pharmacokinetics and exposure-overall survival analysis of the transforming growth factor-β inhibitor galunisertib in patients with pancreatic cancer. Cancer Chemother Pharmacol 2019;84:1003–15. 10.1007/s00280-019-03931-1
    1. Principe DR, Park A, Dorman MJ, et al. . TGFβ blockade augments PD-1 inhibition to promote T-cell-mediated regression of pancreatic cancer. Mol Cancer Ther 2019;18:613–20. 10.1158/1535-7163.MCT-18-0850
    1. Sow HS, Ren J, Camps M, et al. . Combined inhibition of TGF-β signaling and the PD-L1 immune checkpoint is differentially effective in tumor models. Cells 2019;8:320. 10.3390/cells8040320
    1. Gueorguieva I, Cleverly AL, Stauber A, et al. . Defining a therapeutic window for the novel TGF-β inhibitor LY2157299 monohydrate based on a pharmacokinetic/pharmacodynamic model. Br J Clin Pharmacol 2014;77:796–807. 10.1111/bcp.12256
    1. Stauber A, Credille K, Truex L. Nonclinical safety evaluation of a transforming growth factor β receptor I kinase inhibitor in Fischer 344 rats and beagle dogs. J Clin Pract 2014;4:196.
    1. Siu LL, Even C, Mesía R, et al. . Safety and efficacy of durvalumab with or without tremelimumab in patients with PD-L1-low/negative recurrent or metastatic HNSCC: the phase 2 CONDOR randomized clinical trial. JAMA Oncol 2019;5:195–203. 10.1001/jamaoncol.2018.4628
    1. Brandes AA, Carpentier AF, Kesari S, et al. . A phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro Oncol 2016;18:1146–56. 10.1093/neuonc/now009
    1. Kovacs RJ, Maldonado G, Azaro A, et al. . Cardiac safety of TGF-β receptor I kinase inhibitor LY2157299 monohydrate in cancer patients in a first-in-human dose study. Cardiovasc Toxicol 2015;15:309–23. 10.1007/s12012-014-9297-4
    1. Baverel PG, Dubois VFS, Jin CY, et al. . Population pharmacokinetics of durvalumab in cancer patients and association with longitudinal biomarkers of disease status. Clin Pharmacol Ther 2018;103:631–42. 10.1002/cpt.982
    1. O’Reilly EM, D-Y O, Dhani N, et al. . Durvalumab with or without tremelimumab for patients with metastatic pancreatic ductal adenocarcinoma: a phase 2 randomized clinical trial. JAMA Oncol 2019;5:1431–8.
    1. Overman M, Javle M, Davis RE, et al. . Randomized phase II study of the Bruton tyrosine kinase inhibitor acalabrutinib, alone or with pembrolizumab in patients with advanced pancreatic cancer. J Immunother Cancer 2020;8:e000587. 10.1136/jitc-2020-000587
    1. Mahalingam D, Wilkinson GA, Eng KH, et al. . Pembrolizumab in combination with the oncolytic virus pelareorep and chemotherapy in patients with advanced pancreatic adenocarcinoma: a phase Ib study. Clin Cancer Res 2020;26:71–81. 10.1158/1078-0432.CCR-19-2078
    1. Singhi AD, George B, Greenbowe JR, et al. . Real-time targeted genome profile analysis of pancreatic ductal adenocarcinomas identifies genetic alterations that might be targeted with existing drugs or used as biomarkers. Gastroenterology 2019;156:2242–53. 10.1053/j.gastro.2019.02.037
    1. Bernard V, Kim DU, San Lucas FA, et al. . Circulating nucleic acids are associated with outcomes of patients with pancreatic cancer. Gastroenterology 2019;156:108–18. 10.1053/j.gastro.2018.09.022
    1. Macherla S, Laks S, Naqash AR, et al. . Emerging role of immune checkpoint blockade in pancreatic cancer. Int J Mol Sci 2018;19:3505. 10.3390/ijms19113505
    1. Signorelli D, Giannatempo P, Grazia G, et al. . Patients selection for immunotherapy in solid tumors: overcome the naïve vision of a single biomarker. Biomed Res Int 2019;2019.10.1155/2019/9056417
    1. Melisi D, Garcia-Carbonero R, Macarulla T, et al. . TGFβ receptor inhibitor galunisertib is linked to inflammation- and remodeling-related proteins in patients with pancreatic cancer. Cancer Chemother Pharmacol 2019;83:975–91. 10.1007/s00280-019-03807-4
    1. Carstens JL, Correa de Sampaio P, Yang D, et al. . Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer. Nat Commun 2017;8:15095. 10.1038/ncomms15095
    1. Knudsen ES, Vail P, Balaji U, et al. . Stratification of pancreatic ductal adenocarcinoma: combinatorial genetic, stromal, and immunologic markers. Clin Cancer Res 2017;23:4429–40. 10.1158/1078-0432.CCR-17-0162
    1. Mariathasan S, Turley SJ, Nickles D, et al. . TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018;554:544–8. 10.1038/nature25501
    1. Tauriello DVF, Palomo-Ponce S, Stork D, et al. . TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018;554:538–43. 10.1038/nature25492
    1. Pu N, Zhao G, Yin H, et al. . CD25 and TGF-β blockade based on predictive integrated immune ratio inhibits tumor growth in pancreatic cancer. J Transl Med 2018;16:294. 10.1186/s12967-018-1673-6
    1. Ohue Y, Nishikawa H. Regulatory T (Treg) cells in cancer: can Treg cells be a new therapeutic target? Cancer Sci 2019;110:2080–9. 10.1111/cas.14069
    1. Holmgaard RB, Schaer DA, Li Y, et al. . Targeting the TGFβ pathway with galunisertib, a TGFβRI small molecule inhibitor, promotes anti-tumor immunity leading to durable, complete responses, as monotherapy and in combination with checkpoint blockade. J Immunother Cancer 2018;6:47. 10.1186/s40425-018-0356-4
    1. Pu N, Lou W, Yu J. PD-1 immunotherapy in pancreatic cancer: current status. J Pancreatol 2019;2:6–10. 10.1097/JP9.0000000000000010

Source: PubMed

3
Prenumerera