Long-Term Outcomes of a Phase I Study With UV1, a Second Generation Telomerase Based Vaccine, in Patients With Advanced Non-Small Cell Lung Cancer

Paal F Brunsvig, Tormod Kyrre Guren, Marta Nyakas, Claudius H Steinfeldt-Reisse, Wenche Rasch, Jon Amund Kyte, Hedvig Vidarsdotter Juul, Steinar Aamdal, Gustav Gaudernack, Else Marit Inderberg, Paal F Brunsvig, Tormod Kyrre Guren, Marta Nyakas, Claudius H Steinfeldt-Reisse, Wenche Rasch, Jon Amund Kyte, Hedvig Vidarsdotter Juul, Steinar Aamdal, Gustav Gaudernack, Else Marit Inderberg

Abstract

Human telomerase reverse transcriptase (hTERT) is a target antigen for cancer immunotherapy in patients with non-small cell lung cancer (NSCLC). We have tested a novel hTERT vaccine, UV1, designed to give high population coverage. UV1 is composed of three synthetic long peptides containing multiple epitopes identified by epitope spreading data from long-term survivors from previous hTERT vaccination trials. Eighteen non-HLA-typed patients with stage III/IV NSCLC with no evidence of progression after prior treatments, were enrolled in a phase I dose-escalation study of UV1 vaccination with GM-CSF as adjuvant, evaluating safety, immune response, and long-term clinical outcome. Treatment with UV1 was well tolerated with no serious adverse events observed. Seventeen patients were evaluable for tumor response; 15 patients had stable disease as best response. The median progression free survival (PFS) was 10.7 months, and the median overall survival (OS) was 28.2 months. The OS at 4 years was 39% (7/18). Five patients are alive (median survival 5.6 years), and none of these are known to have received checkpoint therapy after vaccination. UV1 induced specific T-cell responses in the majority (67%) of patients. Immune responses were dynamic and long lasting. Both immune response (IR) and OS were dose related. More patients in the highest UV1 dosage group (700 μg) developed IRs compared to the other groups, and the IRs were stronger and occurred earlier. Patients in this group had a 4-year OS of 83%. The safety and clinical outcome data favor 700 μg as the preferred UV1 dose in this patient population. These results provide a rationale for further clinical studies in NSCLC with UV1 vaccination in combination with immune checkpoint blockade.

Clinical trial registration: https://www.clinicaltrials.gov, identifier NCT0178909.

Trial registration: ClinicalTrials.gov NCT01789099.

Keywords: clinical efficacy; dose response; human telomerase reverse transcriptase; non-small cell lung cancer; peptide; vaccine monotherapy.

Conflict of interest statement

GG and EMI are inventors on the UV1 vaccine patent. WR, GG, and EMI are shareholders of Ultimovacs ASA. WR, GG, and SA are Ultimovacs ASA employees. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2020 Brunsvig, Guren, Nyakas, Steinfeldt-Reisse, Rasch, Kyte, Juul, Aamdal, Gaudernack and Inderberg.

Figures

Figure 1
Figure 1
Summary of pre- and post-vaccination UV1-specific T-cell responses. T-cell proliferation against UV1 peptides in pre- and post-vaccination blood samples from the 18 patients evaluable for immune responses. The graph shows the strongest post-vaccination T-cell responses detected against the UV1 peptide mix for each patient (A). Proliferation was measured in response to peptide-loaded PBMC by 3H-thymidine incorporation. A stimulation index (SI) of >3 is considered as an immune response. Stimulation index is plotted as mean of triplicates +/- standard deviation. Twelve of eighteen patients (12/18) had an immune response (67%). In blue: 100µg dose group, in red: 300µg dose group and in green: 700µg dose group (light: pre-vaccination, dark: post-vaccination). T-cell responses against the UV1 vaccine mix and its individual peptide components (725, 728, and 719-20) at all time points measured in patients 908, 700 µg dose (B), 909, 700µg dose (C), 911, 100µg dose (D), and 916, 300µg dose (E). Red arrows indicate when the last vaccine dose was given (B–E).
Figure 2
Figure 2
High UV1 dose improves immune response which correlates with improved OS. The number of patients responding to one (or only UV1 mix), two or three, or no peptides is shown in (A). In (B), the survival (weeks) is shown for patients responding to no peptides, one peptide or two or more. The time-to-response and accumulated proportion of immune responders per UV1 dose group is shown in (C). Overall survival (OS) in immune responders versus non-immune responders is shown in (D).
Figure 3
Figure 3
Tumor reduction and Th1 cytokine production post vaccination. Timeline for diagnosis, treatment and disease progression in patient 908 (A). Summary of pre- and post-vaccination UV1-specific T-cell responses detected, and tumor reduction measured in patient 908 (B). T-cell proliferation against UV1 peptides in blood samples pre- and post-vaccination at all time points tested from patient 908. Proliferation was measured in response to peptide-loaded PBMC by 3H-thymidine incorporation. A stimulation index (SI) of ≥3 is considered as an immune response. Dotted line is the logarithmic trend line for tumor diameter. Th1 cytokine production in T cells stimulated with UV1 peptides post vaccination is shown in (C). TNF-α and IFN-γ production was measured by intracellular cytokine staining in PBMCs pre-stimulated with UV1 peptides. T cells from indicated time points from two patients were re-stimulated with autologous APCs, either PBMCs or B-cell lines (EBV-LCLs) loaded with UV1 peptides (10 µM) for 10 h. CD4+ T-cell response is indicated by black bars and CD8+ T-cell response by hatched bars.
Figure 4
Figure 4
Survival versus months since first UV1 dose. Overall survival is shown in (A) and progression free survival is shown in (B).

References

    1. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Pineros M, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer (2019) 144:1941–53. 10.1002/ijc.31937
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med (2012) 366:2443–54. 10.1056/NEJMoa1200690
    1. Zuazo M, Arasanz H, Fernandez-Hinojal G, Garcia-Granda MJ, Gato M, Bocanegra A, et al. Functional systemic CD4 immunity is required for clinical responses to PD-L1/PD-1 blockade therapy. EMBO Mol Med (2019) 11:e10293. 10.15252/emmm.201910293
    1. Harley CB, Kim NW, Prowse KR, Weinrich SL, Hirsch KS, West MD, et al. Telomerase, cell immortality, and cancer. Cold Spring Harb Symp Quant Biol (1994) 59:307–15. 10.1101/SQB.1994.059.01.035
    1. Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, et al. Specific association of human telomerase activity with immortal cells and cancer. Science (1994) 266:2011–5. 10.1126/science.7605428
    1. Zanetti M. A second chance for telomerase reverse transcriptase in anticancer immunotherapy. Nat Rev Clin Oncol (2017) 14:115–28. 10.1038/nrclinonc.2016.67
    1. Mizukoshi E, Kaneko S. Telomerase-Targeted Cancer Immunotherapy. Int J Mol Sci (2019) 20(8):1823. 10.3390/ijms20081823
    1. Teixeira L, Medioni J, Garibal J, Adotevi O, Doucet L, Dragon Durey MA, et al. A First-in-Human Phase I Study of INVAC-1, an Optimized Human Telomerase DNA Vaccine in Patients with Advanced Solid Tumors. Clin Cancer Res (2020) 26(3):588–97. 10.1158/1078-0432.CCR-19-1614
    1. Wright WE, Piatyszek MA, Rainey WE, Byrd W, Shay JW. Telomerase activity in human germline and embryonic tissues and cells. Dev Genet (1996) 18:173–9. 10.1002/(sici)1520-6408(1996)18:2<173::aid-dvg10>;2-3
    1. Tahara H, Yasui W, Tahara E, Fujimoto J, Ito K, Tamai K, et al. Immuno-histochemical detection of human telomerase catalytic component, hTERT, in human colorectal tumor and non-tumor tissue sections. Oncogene (1999) 18:1561–7. 10.1038/sj.onc.1202458
    1. Brunsvig PF, Aamdal S, Gjertsen MK, Kvalheim G, Markowski-Grimsrud CJ, Sve I, et al. Telomerase peptide vaccination: a phase I/II study in patients with non-small cell lung cancer. Cancer Immunol Immunother : CII (2006) 55:1553–64. 10.1007/s00262-006-0145-7
    1. Danet-Desnoyers G-AH, Luongo JL, Bonnet DA, Domchek SM, Vonderheide RH. Telomerase vaccination has no detectable effect on SCID-repopulating and colony-forming activities in the bone marrow of cancer patients. Exp Hematol (2005) 33:1275–80. 10.1016/j.exphem.2005.07.011
    1. Inderberg-Suso EM, Trachsel S, Lislerud K, Rasmussen AM, Gaudernack G. Widespread CD4+ T-cell reactivity to novel hTERT epitopes following vaccination of cancer patients with a single hTERT peptide GV1001. Oncoimmunology (2012) 1:670–86. 10.4161/onci.20426
    1. Lilleby W, Gaudernack G, Brunsvig PF, Vlatkovic L, Schulz M, Mills K, et al. Phase I/IIa clinical trial of a novel hTERT peptide vaccine in men with metastatic hormone-naive prostate cancer. Cancer Immunol Immunother (2017) 66:891–901. 10.1007/s00262-017-1994-y
    1. Borst J, Ahrends T, Babala N, Melief CJM, Kastenmuller W. CD4(+) T cell help in cancer immunology and immunotherapy. Nat Rev Immunol (2018) 18:635–47. 10.1038/s41577-018-0044-0
    1. Wang L, Soria JC, Kemp BL, Liu DD, Mao L, Khuri FR. hTERT expression is a prognostic factor of survival in patients with stage I non-small cell lung cancer. Clin Cancer Res (2002) 8:2883–9. 10.1093/jnci/93.16.1257
    1. Zhu CQ, Cutz JC, Liu N, Lau D, Shepherd FA, Squire JA, et al. Amplification of telomerase (hTERT) gene is a poor prognostic marker in non-small-cell lung cancer. Br J Cancer (2006) 94:1452–9. 10.1038/sj.bjc.6603110
    1. Jung SJ, Kim DS, Park WJ, Lee H, Choi IJ, Park JY, et al. Mutation of the TERT promoter leads to poor prognosis of patients with non-small cell lung cancer. Oncol Lett (2017) 14:1609–14. 10.3892/ol.2017.6284
    1. Godet Y, Fabre E, Dosset M, Lamuraglia M, Levionnois E, Ravel P, et al. Analysis of spontaneous tumor-specific CD4 T-cell immunity in lung cancer using promiscuous HLA-DR telomerase-derived epitopes: potential synergistic effect with chemotherapy response. Clin Cancer Res (2012) 18:2943–53. 10.1158/1078-0432.CCR-11-3185
    1. Brunsvig PF, Kyte JA, Kersten C, Sundstrom S, Moller M, Nyakas M, et al. Telomerase peptide vaccination in NSCLC: a phase II trial in stage III patients vaccinated after chemoradiotherapy and an 8-year update on a phase I/II trial. Clin Cancer Res (2011) 17:6847–57. 10.1158/1078-0432.CCR-11-1385
    1. Laheurte C, Dosset M, Vernerey D, Boullerot L, Gaugler B, Gravelin E, et al. Distinct prognostic value of circulating anti-telomerase CD4+ Th1 immunity and exhausted PD-1+/TIM-3+ T cells in lung cancer. Br J Cancer (2019) 121:405–16. 10.1038/s41416-019-0531-5
    1. Charo J, Perez C, Buschow C, Jukica A, Czeh M, Blankenstein T. Visualizing the dynamic of adoptively transferred T cells during the rejection of large established tumors. Eur J Immunol (2011) 41:3187–97. 10.1002/eji.201141452
    1. Kyte JA, Gaudernack G, Dueland S, Trachsel S, Julsrud L, Aamdal S. Telomerase peptide vaccination combined with temozolomide: a clinical trial in stage IV melanoma patients. Clin Cancer Res (2011) 17:4568–80. 10.1158/1078-0432.CCR-11-0184
    1. Spitzer MH, Carmi Y, Reticker-Flynn NE, Kwek SS, Madhireddy D, Martins MM, et al. Systemic Immunity Is Required for Effective Cancer Immunotherapy. Cell (2017) 168:487–502.e15. 10.1016/j.cell.2016.12.022
    1. Gandhi L, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N Engl J Med (2018) 378:2078–92. 10.1056/NEJMoa1801005
    1. Arakawa A, Vollmer S, Tietze J, Galinski A, Heppt MV, Burdek M, et al. Clonality of CD4(+) Blood T Cells Predicts Longer Survival With CTLA4 or PD-1 Checkpoint Inhibition in Advanced Melanoma. Front Immunol (2019) 10:1336. 10.3389/fimmu.2019.01336
    1. Johnson AM, Bullock BL, Neuwelt AJ, Poczobutt JM, Kaspar RE, Li HY, et al. Cancer Cell–Intrinsic Expression of MHC Class II Regulates the Immune Microenvironment and Response to Anti–PD-1 Therapy in Lung Adenocarcinoma. J Immunol (2020) 204:2295–307. 10.4049/jimmunol.1900778
    1. Wada S, Jackson CM, Yoshimura K, Yen HR, Getnet D, Harris TJ, et al. Sequencing CTLA-4 blockade with cell-based immunotherapy for prostate cancer. J Transl Med (2013) 11:89. 10.1186/1479-5876-11-89
    1. Soares KC, Rucki AA, Wu AA, Olino K, Xiao Q, Chai Y, et al. PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors. J Immunother (2015) 38:1–11. 10.1097/CJI.0000000000000062
    1. Massarelli E, William W, Johnson F, Kies M, Ferrarotto R, Guo M, et al. Combining Immune Checkpoint Blockade and Tumor-Specific Vaccine for Patients With Incurable Human Papillomavirus 16-Related Cancer: A Phase 2 Clinical Trial. JAMA Oncol (2019) 5:67–73. 10.1001/jamaoncol.2018.4051
    1. Curran MA, Glisson BS. New Hope for Therapeutic Cancer Vaccines in the Era of Immune Checkpoint Modulation. Annu Rev Med (2019) 70:409–24. 10.1146/annurev-med-050217-121900
    1. Wei SC, Sharma R, Anang NAS, Levine JH, Zhao Y, Mancuso JJ, et al. Negative Co-stimulation Constrains T Cell Differentiation by Imposing Boundaries on Possible Cell States. Immunity (2019) 50:1084–98.e10. 10.1016/j.immuni.2019.03.004
    1. Wei SC, Levine JH, Cogdill AP, Zhao Y, Anang NAS, Andrews MC, et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell (2017) 170:1120–33.e17. 10.1016/j.cell.2017.07.024

Source: PubMed

3
Prenumerera