Clinical and Immune Effects of Lenalidomide in Combination with Gemcitabine in Patients with Advanced Pancreatic Cancer

Gustav J Ullenhag, Fariba Mozaffari, Mats Broberg, Håkan Mellstedt, Maria Liljefors, Gustav J Ullenhag, Fariba Mozaffari, Mats Broberg, Håkan Mellstedt, Maria Liljefors

Abstract

Purpose: To assess the immunomodulatory and clinical effects of lenalidomide with standard treatment of gemcitabine in patients with advanced pancreatic cancer.

Patients and methods: Patients with advanced pancreatic cancer were treated in first line with lenalidomide orally for 21 days of a 28 days cycle and the standard regimen for gemcitabine. In Part I, which we previously have reported, the dose of lenalidomide was defined (n = 12). In Part II, every other consecutive patient was treated with either lenalidomide (Group A, n = 11) or gemcitabine (Group B, n = 10) during cycle 1. From cycle 2 on, all Part II patients received the combination.

Results: A significant decrease in the proliferative response of peripheral blood mononuclear cells and the frequency of DCs were noted in patients at baseline compared to healthy control donors while the frequencies of CD4+ and CD8+ T cells, NK-cells and MDSCs were significantly higher in patients compared to controls. In Group A, a significant increase in the absolute numbers of activated (HLA-DR+) CD4 and CD8 T cells and CD8 effector memory T cells (p<0.01) was noted during treatment. A statistical increment in the absolute numbers of Tregs were seen after cycle 1 (p<0.05). The addition of gemcitabine, reduced most lymphocyte subsets (p<0.05). In Group B, the proportion of lymphocytes remained unchanged during the study period. There was no difference in overall survival, progression free survival and survival rate at one year comparing the two groups.

Discussion: Patients with advanced pancreatic carcinoma had impaired immune functions. Lenalidomide augmented T cell reactivities, which were abrogated by gemcitabine. However, addition of lenalidomide to gemcitabine seemed to have no therapeutic impact compared to gemcitabine alone in this non-randomized study.

Trial registration: ClinicalTrials.gov NCT01547260.

Conflict of interest statement

I have read the journal's policy and one author of this manuscript has the following competing interest. Maria Liljefors has declared one compensatory advisory role with Celgene Corporation. This does not alter the authors adherence to PLoS One policies on sharing data and materials. The other authors have declared that no competing interests exist.

Figures

Fig 1. CONSORT Flow Diagram for the…
Fig 1. CONSORT Flow Diagram for the patients screened for and enrolled in part II.
Corresponding data for patients enrolled in part I, has been published in Ullenhag GJ et al, PLOS ONE, 2015; 10(4).
Fig 2
Fig 2
Absolute numbers of subsets of T cells at baseline (BL), at the end of cycle 1 (End C1) and at the end of cycle 2 (End C2) in part II patients treated with either lenalidomide monotherapy during cycle 1 with the addition of gemcitabine from cycle 2 (Arm A) (Left column) (Fig 2 A, C, E and G) or gemcitabine monotherapy during cycle 1 with the addition of lenalidomide from cycle 2 (Arm B) (Right column) (Fig 2 B, D, F and H). Changes in the absolute numbers of CD4+ T cells (A, B), CD8+ T cells (C, D), HLA-DR positive CD+4 T cells (E, F) and HLA-DR positive CD8+ T cells (G, H) over the treatment course, n = number of patients analysed at each time-point. P-values refer to the comparison with BL, End C1 and End C2 by one way ANOVA with repeated measures. The box, with a line indicating median, represents the 25th and 75th percentiles. The top and bottom whiskers represent the 90th and 10th percentiles, respectively.
Fig 3
Fig 3
Absolute numbers of subsets of effector and effector memory T cells at baseline (BL), at the end of cycle 1 (End C1) and at the end of cycle 2 (End C2) in part II patients treated with either lenalidomide monotherapy during cycle 1 with the addition of gemcitabine from cycle 2 (Arm A) (Left column) (Fig 3 A, C, E and G) or gemcitabine monotherapy during cycle 1 with the addition of lenalidomide from cycle 2 (Arm B) (Right column) (Fig 3 B, D, F and H). Changes in the absolute numbers of CD8+ effector T cells (CD45RA+CCR7-CD8+) (C, D), CD8+ effector memory T cells (CD45RA-CCR7-CD8+) (G, H), CD4+ effector T cells (CD45RA+CCR7-CD4+) (A-B) and CD4+ effector memory T cells (CD45RA-CCR7-CD4+) (E, F) over the treatment course. n = number of patients analysed at each time-point. P-values refer to the comparison with BL, End C1 and End C2 by one way ANOVA with repeated measures. The box, with a line indicating median, represents the 25th and 75th percentiles. The top and bottom whiskers represent the 90th and 10th percentiles, respectively.
Fig 4
Fig 4
Absolute numbers of NKT-cells (CD3+CD56+CD16+) and regulatory T cells (Treg)(CD4+CD25+CD127-Foxp3+) at baseline (BL), at the end of cycle 1 (End C1) and at the end of cycle 2 (End C2) in part II patients treated with either lenalidomide monotherapy during cycle 1 with the addition of gemcitabine from cycle 2 (Arm A) (Left column) (Fig 4 A and C) or gemcitabine monotherapy during cycle 1 with the addition of lenalidomide from cycle 2 (Arm B) (Right column) (Fig 4 B and D). Changes in the absolute numbers of NKT-cells (A, B) and regulatory T cells (C, D) over the treatment course. n = number of patients analysed at each time-point. P-values refer to the comparison with BL, End C1 and End C2 by one way ANOVA with repeated measures. The box, with a line indicating median, represents the 25th and 75th percentiles. The top and bottom whiskers represent the 90th and 10th percentiles, respectively.
Fig 5. Cumulative overall survival (OS) (solid…
Fig 5. Cumulative overall survival (OS) (solid black line) and progression-free survival (PFS) (solid grey line) from the start of study treatment until death (OS) and clinical and/or radiological signs of disease progression (PFS), respectively, for all patients in the study.
Number of evaluable patients for OS = 33 out of 33, for PFS = 29 out of 33.

References

    1. Alexakis N, Halloran C, Raraty M, Ghaneh P, Sutton R, Neoptolemos JP. Current standards of surgery for pancreatic cancer. Br J Surg. 2004;91(11):1410–27. 10.1002/bjs.4794
    1. Freelove R, Walling AD. Pancreatic cancer: diagnosis and management. Am Fam Physician. 2006;73(3):485–92.
    1. Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. Jama. 2010;304(10):1073–81. 10.1001/jama.2010.1275
    1. Burris HA 3rd, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15(6):2403–13. 10.1200/jco.1997.15.6.2403
    1. Cunningham D, Chau I, Stocken DD, Valle JW, Smith D, Steward W, et al. Phase III randomized comparison of gemcitabine versus gemcitabine plus capecitabine in patients with advanced pancreatic cancer. J Clin Oncol. 2009;27(33):5513–8. 10.1200/JCO.2009.24.2446
    1. Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. The New England journal of medicine. 2011;364(19):1817–25. 10.1056/NEJMoa1011923
    1. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25(15):1960–6. 10.1200/JCO.2006.07.9525
    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. The New England journal of medicine. 2013;369(18):1691–703. PubMed Central PMCID: PMC4631139. 10.1056/NEJMoa1304369
    1. FDA US Food and Drug Administration (FDA) 2013. Available from: .
    1. European Medicine Agency (EMA) Science Medicine Health. SPC_Lenalidomide_PDF.REVLIMID—revised Feb 2013 2013. Available from: .
    1. Dredge K, Horsfall R, Robinson SP, Zhang LH, Lu L, Tang Y, et al. Orally administered lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits endothelial cell migration and Akt phosphorylation in vitro. Microvasc Res. 2005;69(1–2):56–63. 10.1016/j.mvr.2005.01.002
    1. Wu L, Parton A, Lu L, Adams M, Schafer P, Bartlett JB. Lenalidomide enhances antibody-dependent cellular cytotoxicity of solid tumor cells in vitro: influence of host immune and tumor markers. Cancer immunology, immunotherapy: CII. 2011;60(1):61–73. 10.1007/s00262-010-0919-9
    1. LeBlanc R, Hideshima T, Catley LP, Shringarpure R, Burger R, Mitsiades N, et al. Immunomodulatory drug costimulates T cells via the B7-CD28 pathway. Blood. 2004;103(5):1787–90. 10.1182/blood-2003-02-0361
    1. Teo SK. Properties of thalidomide and its analogues: implications for anticancer therapy. AAPS J. 2005;7(1):E14–9. PubMed Central PMCID: PMC2751493. 10.1208/aapsj070103
    1. Corral LG, Haslett PA, Muller GW, Chen R, Wong LM, Ocampo CJ, et al. Differential cytokine modulation and T cell activation by two distinct classes of thalidomide analogues that are potent inhibitors of TNF-alpha. J Immunol. 1999;163(1):380–6.
    1. Crane E, List A. Immunomodulatory drugs. Cancer Invest. 2005;23(7):625–34. 10.1080/07357900500283101
    1. Amato RJ, Hernandez-McClain J, Saxena S, Khan M. Lenalidomide therapy for metastatic renal cell carcinoma. Am J Clin Oncol. 2008;31(3):244–9. 10.1097/COC.0b013e31815e451f
    1. Bartlett JB, Michael A, Clarke IA, Dredge K, Nicholson S, Kristeleit H, et al. Phase I study to determine the safety, tolerability and immunostimulatory activity of thalidomide analogue CC-5013 in patients with metastatic malignant melanoma and other advanced cancers. Br J Cancer. 2004;90(5):955–61. PubMed Central PMCID: PMC2410215. 10.1038/sj.bjc.6601579
    1. Miller AA, Case D, Harmon M, Savage P, Lesser G, Hurd D, et al. Phase I study of lenalidomide in solid tumors. J Thorac Oncol. 2007;2(5):445–9. 10.1097/01.JTO.0000268679.33238.67
    1. Sharma RA, Steward WP, Daines CA, Knight RD, O'Byrne KJ, Dalgleish AG. Toxicity profile of the immunomodulatory thalidomide analogue, lenalidomide: phase I clinical trial of three dosing schedules in patients with solid malignancies. Eur J Cancer. 2006;42(14):2318–25. 10.1016/j.ejca.2006.05.018
    1. Sanborn SL, Gibbons J, Krishnamurthi S, Brell JM, Dowlati A, Bokar JA, et al. Phase I trial of docetaxel given every 3 weeks and daily lenalidomide in patients with advanced solid tumors. Invest New Drugs. 2009;27(5):453–60. 10.1007/s10637-008-9200-x
    1. Fryer RA, Barlett B, Galustian C, Dalgleish AG. Mechanisms underlying gemcitabine resistance in pancreatic cancer and sensitisation by the iMiD lenalidomide. Anticancer research. 2011;31(11):3747–56.
    1. Fritz S, Hackert T, Buchler MW. Pancreatic intraductal papillary mucinous neoplasm—where is the challenge? Digestive diseases. 2015;33(1):99–105. 10.1159/000368448
    1. Suzuki E, Sun J, Kapoor V, Jassar AS, Albelda SM. Gemcitabine has significant immunomodulatory activity in murine tumor models independent of its cytotoxic effects. Cancer biology & therapy. 2007;6(6):880–5.
    1. Plate JM, Plate AE, Shott S, Bograd S, Harris JE. Effect of gemcitabine on immune cells in subjects with adenocarcinoma of the pancreas. Cancer immunology, immunotherapy: CII. 2005;54(9):915–25. 10.1007/s00262-004-0638-1
    1. Soeda A, Morita-Hoshi Y, Makiyama H, Morizane C, Ueno H, Ikeda M, et al. Regular dose of gemcitabine induces an increase in CD14+ monocytes and CD11c+ dendritic cells in patients with advanced pancreatic cancer. Japanese journal of clinical oncology. 2009;39(12):797–806. 10.1093/jjco/hyp112
    1. Nowak AK, Lake RA, Marzo AL, Scott B, Heath WR, Collins EJ, et al. Induction of tumor cell apoptosis in vivo increases tumor antigen cross-presentation, cross-priming rather than cross-tolerizing host tumor-specific CD8 T cells. J Immunol. 2003;170(10):4905–13.
    1. Homma Y, Taniguchi K, Nakazawa M, Matsuyama R, Mori R, Takeda K, et al. Changes in the immune cell population and cell proliferation in peripheral blood after gemcitabine-based chemotherapy for pancreatic cancer. Clinical & translational oncology: official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico. 2014;16(3):330–5.
    1. Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clinical cancer research: an official journal of the American Association for Cancer Research. 2005;11(18):6713–21.
    1. Vizio B, Novarino A, Giacobino A, Cristiano C, Prati A, Ciuffreda L, et al. Potential plasticity of T regulatory cells in pancreatic carcinoma in relation to disease progression and outcome. Experimental and therapeutic medicine. 2012;4(1):70–8. PubMed Central PMCID: PMC3460315. 10.3892/etm.2012.553
    1. Bergmann-Leitner ES, Abrams SI. Treatment of human colon carcinoma cell lines with anti-neoplastic agents enhances their lytic sensitivity to antigen-specific CD8+ cytotoxic T lymphocytes. Cancer immunology, immunotherapy: CII. 2001;50(9):445–55.
    1. Ullenhag GJ, Rossmann E, Liljefors M. A phase I dose-escalation study of lenalidomide in combination with gemcitabine in patients with advanced pancreatic cancer. PloS one. 2015;10(4):e0121197 PubMed Central PMCID: PMC4383423. 10.1371/journal.pone.0121197
    1. Mozaffari F, Lindemalm C, Choudhury A, Granstam-Bjorneklett H, Lekander M, Nilsson B, et al. Systemic immune effects of adjuvant chemotherapy with 5-fluorouracil, epirubicin and cyclophosphamide and/or radiotherapy in breast cancer: a longitudinal study. Cancer immunology, immunotherapy: CII. 2009;58(1):111–20. 10.1007/s00262-008-0530-5
    1. Lindemalm C, Mozaffari F, Choudhury A, Granstam-Bjorneklett H, Lekander M, Nilsson B, et al. Immune response, depression and fatigue in relation to support intervention in mammary cancer patients. Supportive care in cancer: official journal of the Multinational Association of Supportive Care in Cancer. 2008;16(1):57–65.
    1. Bazhin AV, Shevchenko I, Umansky V, Werner J, Karakhanova S. Two immune faces of pancreatic adenocarcinoma: possible implication for immunotherapy. Cancer immunology, immunotherapy: CII. 2014;63(1):59–65. 10.1007/s00262-013-1485-8
    1. Yanagimoto H, Takai S, Satoi S, Toyokawa H, Takahashi K, Terakawa N, et al. Impaired function of circulating dendritic cells in patients with pancreatic cancer. Clinical immunology. 2005;114(1):52–60. 10.1016/j.clim.2004.09.007
    1. Yamamoto T, Yanagimoto H, Satoi S, Toyokawa H, Hirooka S, Yamaki S, et al. Circulating CD4+CD25+ regulatory T cells in patients with pancreatic cancer. Pancreas. 2012;41(3):409–15. 10.1097/MPA.0b013e3182373a66
    1. Schmitz-Winnenthal FH, Volk C, Z'Graggen K, Galindo L, Nummer D, Ziouta Y, et al. High frequencies of functional tumor-reactive T cells in bone marrow and blood of pancreatic cancer patients. Cancer research. 2005;65(21):10079–87. 10.1158/0008-5472.CAN-05-1098
    1. Pituch-Noworolska A. The response of cancer patients' lymphocytes to mitogen. The effect of indomethacin on adherent and non-adherent cells activity. Archivum immunologiae et therapiae experimentalis. 1981;29(2):161–5.
    1. Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer research. 2010;70(1):68–77. PubMed Central PMCID: PMC2805057. 10.1158/0008-5472.CAN-09-2587
    1. Wolf AM, Wolf D, Steurer M, Gastl G, Gunsilius E, Grubeck-Loebenstein B. Increase of regulatory T cells in the peripheral blood of cancer patients. Clinical cancer research: an official journal of the American Association for Cancer Research. 2003;9(2):606–12.
    1. Iakoucheva LM, Brown CJ, Lawson JD, Obradovic Z, Dunker AK. Intrinsic disorder in cell-signaling and cancer-associated proteins. Journal of molecular biology. 2002;323(3):573–84.
    1. Lioznov M, El-Cheikh J Jr., Hoffmann F, Hildebrandt Y, Ayuk F, Wolschke C, et al. Lenalidomide as salvage therapy after allo-SCT for multiple myeloma is effective and leads to an increase of activated NK (NKp44(+)) and T (HLA-DR(+)) cells. Bone marrow transplantation. 2010;45(2):349–53. 10.1038/bmt.2009.155
    1. Galustian C, Meyer B, Labarthe MC, Dredge K, Klaschka D, Henry J, et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer immunology, immunotherapy: CII. 2009;58(7):1033–45. 10.1007/s00262-008-0620-4
    1. Lee BN, Gao H, Cohen EN, Badoux X, Wierda WG, Estrov Z, et al. Treatment with lenalidomide modulates T-cell immunophenotype and cytokine production in patients with chronic lymphocytic leukemia. Cancer. 2011;117(17):3999–4008. PubMed Central PMCID: PMC4349201. 10.1002/cncr.25983
    1. Hayashi T, Hideshima T, Akiyama M, Podar K, Yasui H, Raje N, et al. Molecular mechanisms whereby immunomodulatory drugs activate natural killer cells: clinical application. British journal of haematology. 2005;128(2):192–203. 10.1111/j.1365-2141.2004.05286.x
    1. Infante JR, Arkenau HT, Bendell JC, Rubin MS, Waterhouse D, Jones GT, et al. Lenalidomide in combination with gemcitabine as first-line treatment for patients with metastatic carcinoma of the pancreas: a Sarah Cannon Research Institute phase II trial. Cancer biology & therapy. 2013;14(4):340–6. PubMed Central PMCID: PMC3667874.
    1. Infante JR, Jones SF, Bendell JC, Spigel DR, Yardley DA, Weekes CD, et al. A phase I, dose-escalation study of pomalidomide (CC-4047) in combination with gemcitabine in metastatic pancreas cancer. Eur J Cancer. 2011;47(2):199–205. 10.1016/j.ejca.2010.09.002
    1. Maraveyas A, Waters J, Roy R, Fyfe D, Propper D, Lofts F, et al. Gemcitabine versus gemcitabine plus dalteparin thromboprophylaxis in pancreatic cancer. Eur J Cancer. 2012;48(9):1283–92. 10.1016/j.ejca.2011.10.017

Source: PubMed

3
Sottoscrivi