A One-Armed Phase I Dose Escalation Trial Design: Personalized Vaccination with IKKβ-Matured, RNA-Loaded Dendritic Cells for Metastatic Uveal Melanoma

Elias A T Koch, Niels Schaft, Mirko Kummer, Carola Berking, Gerold Schuler, Kenichiro Hasumi, Jan Dörrie, Beatrice Schuler-Thurner, Elias A T Koch, Niels Schaft, Mirko Kummer, Carola Berking, Gerold Schuler, Kenichiro Hasumi, Jan Dörrie, Beatrice Schuler-Thurner

Abstract

Uveal melanoma (UM) is an orphan disease with a mortality of 80% within one year upon the development of metastatic disease. UM does hardly respond to chemotherapy and kinase inhibitors and is largely resistant to checkpoint inhibition. Hence, further therapy approaches are urgently needed. To improve clinical outcome, we designed a trial employing the 3rd generation personalized IKKβ-matured RNA-transfected dendritic cell (DC) vaccine which primes T cells and in addition activates NK cells. This ongoing phase I trial [NCT04335890 (www.clinicaltrials.gov), Eudract: 2018-004390-28 (www.clinicaltrialsregister.eu)] investigates patients with treatment-naive metastatic UM. Monocytes are isolated by leukapheresis, differentiated to immature DCs, matured with a cytokine cocktail, and activated via the NF-κB pathway by electroporation with RNA encoding a constitutively active mutant of IKKβ. Three types of antigen-RNA are co-electroporated: i) amplified mRNA of the tumor representing the whole transcriptome, ii) RNA encoding driver mutations identified by exome sequencing, and iii) overexpressed non-mutated tumor antigens detected by transcriptome sequencing. This highly personalized DC vaccine is applied by 9 intravenous infusions in a staggered schedule over one year. Parallel to the vaccination, standard therapy, usually an immune checkpoint blockade (ICB) as mono (anti-PD-1) or combined (anti-CTLA4 and anti-PD-1) regimen is initiated. The coordinated vaccine-induced immune response encompassing tumor-specific T cells and innate NK cells should synergize with ICB, perhaps resulting in measurable clinical responses in this resistant tumor entity. Primary outcome measures of this trial are safety, tolerability and toxicity; secondary outcome measures comprise overall survival and induction of antigen-specific T cells.

Keywords: IKKβ-matured dendritic cells; Immune checkpoint blockade; metastatic uveal melanoma; personalized vaccine; tumor antigen vaccine; tumor antigens.

Conflict of interest statement

The authors declare the following potential conflict of interest: GS, NS, and JD are named as inventors on a patent on caIKK-RNA-electroporated DCs (WO/2012/055551), which is held by the Friedrich-Alexander-Universtät Erlangen-Nürnberg. 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 © 2022 Koch, Schaft, Kummer, Berking, Schuler, Hasumi, Dörrie and Schuler-Thurner.

Figures

Figure 1
Figure 1
DCs present tumor antigens through MHC class I and II to CD8+ and CD4+ T cells, respectively, and are able to activate innate immune cells, like NK cells. T-cell priming is supported by co-stimulatory molecules and the secretion of pro-inflammatory cytokines. NF-κB-activated DCs express increased levels of the classical co-stimulatory molecules CD80 and CD86, but also of CD70, CD40, OX40L, and several pro-inflammatory cytokines, such as IL-12 and TNFα, which support induction of memory T-cell responses and increase NK-cell activation and proliferation. The antigens used in this trial are modified in such a way, that both MHC class I- and class II-restricted presentation is facilitated. Therefore, a comprehensive cellular response of helper T cells, cytotoxic T cells, and NK cells is induced, which in turn produce additional cytokines that support activation.
Figure 2
Figure 2
Treatment schedule: Initially a tumor biopsy is taken, and RNA and DNA preparation and sequencing is performed. To obtain the required numbers of autologous DCs, a monocyte concentrate is collected via leukapheresis. The vaccine is generated by RNA-electroporation of DCs and 9 vaccine injections are given intravenously over a period of one year. Every three months CT/MRI staging is performed.

References

    1. Baily C, O'Neill V, Dunne M, Cunningham M, Gullo G, Kennedy S, et al. . Uveal Melanoma in Ireland. Ocul Oncol Pathol (2019) 5(3):195–204. doi: 10.1159/000492391
    1. Virgili G, Gatta G, Ciccolallo L, Capocaccia R, Biggeri A, Crocetti E, et al. . Incidence of Uveal Melanoma in Europe. Ophthalmology (2007) 114(12):2309–15. doi: 10.1016/j.ophtha.2007.01.032
    1. Singh AD, Turell ME, Topham AK. Uveal Melanoma: Trends in Incidence, Treatment, and Survival. Ophthalmology (2011) 118(9):1881–5. doi: 10.1016/j.ophtha.2011.01.040
    1. Sayan M, Mamidanna S, Oncel D, Jan I, Vergalasova I, Weiner J, et al. . Clinical Management of Uveal Melanoma: A Comprehensive Review With a Treatment Algorithm. Radiat Oncol J (2020) 38(3):162–9. doi: 10.3857/roj.2020.00318
    1. Fili M, Trocme E, Bergman L, See TRO, André H, Bartuma K, et al. . Ruthenium-106 Versus Iodine-125 Plaque Brachytherapy of 571 Choroidal Melanomas With a Thickness of >/=5.5 Mm. Br J Ophthalmol (2020) 104(1):26–32. doi: 10.1136/bjophthalmol-2018-313419
    1. Collaborative Ocular Melanoma Study G . Assessment of Metastatic Disease Status at Death in 435 Patients With Large Choroidal Melanoma in the Collaborative Ocular Melanoma Study (COMS): COMS Report No. 15. Arch Ophthalmol (2001) 119(5):670–6. doi: 10.1001/archopht.119.5.670
    1. Kujala E, Makitie T, Kivela T. Very Long-Term Prognosis of Patients With Malignant Uveal Melanoma. Invest Ophthalmol Vis Sci (2003) 44(11):4651–9. doi: 10.1167/iovs.03-0538
    1. Franklin C, Livingstone E, Roesch A, Schilling B, Schadendorf D. Immunotherapy in Melanoma: Recent Advances and Future Directions. Eur J Surg Oncol (2017) 43(3):604–11. doi: 10.1016/j.ejso.2016.07.145
    1. Khoja L, Atenafu EG, Suciu S, Leyvraz S, Sato T, Marshall E, et al. . Meta-Analysis in Metastatic Uveal Melanoma to Determine Progression Free and Overall Survival Benchmarks: An International Rare Cancers Initiative (IRCI) Ocular Melanoma Study. Ann Oncol (2019) 30(8):1370–80. doi: 10.1093/annonc/mdz176
    1. Pelster MS, Gruschkus SK, Bassett R, Gombos DS, Shephard M, Posada L, et al. . Nivolumab and Ipilimumab in Metastatic Uveal Melanoma: Results From a Single-Arm Phase II Study. J Clin Oncol (2020) 39(6):599–607. doi: 10.1200/JCO.20.00605
    1. Piulats JM, Espinosa E, de la Cruz Merino L, Varela M, Carrión LA, Martín-Algarra S, et al. . Nivolumab Plus Ipilimumab for Treatment-Naive Metastatic Uveal Melanoma: An Open-Label, Multicenter, Phase II Trial by the Spanish Multidisciplinary Melanoma Group (GEM-1402). J Clin Oncol (2021) 119(5):670–6. doi: 10.1200/JCO.20.00550
    1. Heppt MV, Amaral T, Kahler KC, Heinzerling L, Hassel JC, Meissner M, et al. . Combined Immune Checkpoint Blockade for Metastatic Uveal Melanoma: A Retrospective, Multi-Center Study. J Immunother Cancer (2019) 7(1):299. doi: 10.1186/s40425-019-0800-0
    1. Heppt MV, Heinzerling L, Kahler KC, Forschner A, Kirchberger MC, Loquai C, et al. . Prognostic Factors and Outcomes in Metastatic Uveal Melanoma Treated With Programmed Cell Death-1 or Combined PD-1/Cytotoxic T-Lymphocyte Antigen-4 Inhibition. Eur J Cancer (2017) 82:56–65. doi: 10.1016/j.ejca.2017.05.038
    1. Najjar YG, Navrazhina K, Ding F, Bhatia R, Tsai K, Abbate K, et al. . Ipilimumab Plus Nivolumab for Patients With Metastatic Uveal Melanoma: A Multicenter, Retrospective Study. J Immunother Cancer (2020) 8(1):e000331. doi: 10.1136/jitc-2019-000331
    1. Koch EAT, Petzold A, Wessely A, Dippel E, Gesierich A, Gutzmer R, et al. . Immune Checkpoint Blockade for Metastatic Uveal Melanoma: Patterns of Response and Survival According to the Presence of Hepatic and Extrahepatic Metastasis. Cancers (2021) 13(13):3359. doi: 10.3390/cancers13133359
    1. Hassel JC, Heinzerling L, Aberle J, Bähr O, Eigentler TK, Grimm MO, et al. . Combined Immune Checkpoint Blockade (Anti-PD-1/Anti-CTLA-4): Evaluation and Management of Adverse Drug Reactions. Cancer Treat Rev (2017) 57:36–49. doi: 10.1016/j.ctrv.2017.05.003
    1. Heinzerling L, Ascierto PA, Dummer R, Gogas H, Grob JJ, Lebbe C, et al. . Adverse Events 2.0-Let Us Get SERIOs: New Reporting for Adverse Event Outcomes Needed in the Era of Immunooncology. Eur J Cancer (2019) 112:29–31. doi: 10.1016/j.ejca.2019.01.015
    1. Koch EAT, Nickel FT, Heinzerling L, Schulz YK, Berking C, Erdmann M. Immune Checkpoint Inhibitor-Induced Bilateral Vestibulopathy. J Immunother (2021) 44(3):114–7. doi: 10.1097/CJI.0000000000000353
    1. Sato T, Nathan PD, Hernandez-Aya L, Sacco JJ, Orloff MM, Visich J, et al. . Redirected T Cell Lysis in Patients With Metastatic Uveal Melanoma With Gp100-Directed TCR IMCgp100: Overall Survival Findings. J Clin Oncol (2018) 36(15_suppl):9521–1. doi: 10.1200/JCO.2018.36.15_suppl.9521
    1. Piperno-Neumann S, Hassel JC, Rutkowski P, Baurain JF, Butler MO, Schlaak M, et al. . Abstract CT002: Phase 3 Randomized Trial Comparing Tebentafusp With Investigator's Choice in First Line Metastatic Uveal Melanoma. Cancer Res (2021) 81(13 Supplement):CT002–2. doi: 10.1158/1538-7445.AM2021-CT002
    1. Moreira A, Gross S, Uslu U, Dörrie J, Kummer M, Schliep S, et al. . Abstract E21024: Dendritic Cell Vaccination in Metastatic Uveal Melanoma as Compassionate Treatment: Immunological and Clinical Responses. J Clin Oncol (2019) 37(15_suppl):e21024. doi: 10.1200/JCO.2019.37.15.15_suppl.e21024
    1. Schuler-Thurner B, Bartz-Schmidt KU, Bornfeld N, Cursiefen C, Fuisting B, Grisanti S, et al. . Immunotherapy of Uveal Melanoma: Vaccination Against Cancer. Multicenter Adjuvant Phase 3 Vaccination Study Using Dendritic Cells Laden With Tumor RNA for Large Newly Diagnosed Uveal Melanoma. Ophthalmologe (2015) 112(12):1017–21. doi: 10.1007/s00347-015-0162-z
    1. Pfeiffer IA, Hoyer S, Gerer KF, Voll RE, Knippertz I, Gückel E, et al. . Triggering of NF-kappaB in Cytokine-Matured Human DCs Generates Superior DCs for T-Cell Priming in Cancer Immunotherapy. Eur J Immunol (2014) 44(11):3413–28. doi: 10.1002/eji.201344417
    1. Bosch NC, Voll RE, Voskens CJ, Gross S, Seliger B, Schuler G, et al. . NF-kappaB Activation Triggers NK-Cell Stimulation by Monocyte-Derived Dendritic Cells. Ther Adv Med Oncol (2019) 11:1758835919891622. doi: 10.1177/1758835919891622
    1. Bonehill A, Heirman C, Tuyaerts S, Michiels A, Breckpot K, Brasseur F, et al. . Messenger RNA-Electroporated Dendritic Cells Presenting MAGE-A3 Simultaneously in HLA Class I and Class II Molecules. J Immunol (2004) 172(11):6649–57. doi: 10.4049/jimmunol.172.11.6649
    1. Erdmann M, Dorrie J, Schaft N, Strasser E, Hendelmeier M, Kämpgen E, et al. . Effective Clinical-Scale Production of Dendritic Cell Vaccines by Monocyte Elutriation Directly in Medium, Subsequent Culture in Bags and Final Antigen Loading Using Peptides or RNA Transfection. J Immunother (2007) 30(6):663–74. doi: 10.1097/CJI.0b013e3180ca7cd6
    1. Gerer KF, Hoyer S, Dorrie J, Schaft N. Electroporation of mRNA as Universal Technology Platform to Transfect a Variety of Primary Cells With Antigens and Functional Proteins. Methods Mol Biol (2017) 1499:165–78. doi: 10.1007/978-1-4939-6481-9_10
    1. Dorrie J, Schaft N, Muller I, Wellner V, Schunder T, Hänig J, et al. . Introduction of Functional Chimeric E/L-Selectin by RNA Electroporation to Target Dendritic Cells From Blood to Lymph Nodes. Cancer Immunol Immunother (2008) 57(4):467–77. doi: 10.1007/s00262-007-0385-1
    1. Gross S, Erdmann M, Haendle I, Voland S, Berger T, Schultz E, et al. . Twelve-Year Survival and Immune Correlates in Dendritic Cell-Vaccinated Melanoma Patients. JCI Insight (2017) 2(8):e91438. doi: 10.1172/jci.insight.91438
    1. Dörrie J, Schaft N, Gross S, Ostalecki C, Eberhardt M, Vera-Gonzáles J, et al. . Step by Step Optimization of mRNA-Electroporated Dendritic Cells During a Phase I/II Vaccine Trial in Stage IV Cutaneous Melanoma Patients: An Increase in Survival Correlates With Higher Immunoscore in Metastases, and Upregulation of PEBP1 in Peripheral Blood. J Dtsch Dermatol Ges (2020) 18(S2):8–9. doi: 10.1111/ddg.14192
    1. De Keersmaecker B, Claerhout S, Carrasco J, Bar I, Coerthals J, Wilgenhof S, et al. . TriMix and Tumor Antigen mRNA Electroporated Dendritic Cell Vaccination Plus Ipilimumab: Link Between T-Cell Activation and Clinical Responses in Advanced Melanoma. J Immunother Cancer (2020) 8(1):e000329. doi: 10.1136/jitc-2019-000329
    1. Dörrie J, Schaft N, Schuler G, Schuler-Thurner B. Therapeutic Cancer Vaccination With Ex Vivo RNA-Transfected Dendritic Cells-An Update. Pharmaceutics (2020) 12(2):92. doi: 10.3390/pharmaceutics12020092
    1. Chen PW, Mellon JK, Mayhew E, Wang S, He YG, Hogan N, et al. . Uveal Melanoma Expression of Indoleamine 2,3-Deoxygenase: Establishment of an Immune Privileged Environment by Tryptophan Depletion. Exp Eye Res (2007) 85(5):617–25. doi: 10.1016/j.exer.2007.07.014
    1. Ryu YH, Kim JC. Expression of Indoleamine 2,3-Dioxygenase in Human Corneal Cells as a Local Immunosuppressive Factor. Invest Ophthalmol Vis Sci (2007) 48(9):4148–52. doi: 10.1167/iovs.05-1336
    1. Bjoern J, Iversen TZ, Nitschke NJ, Andersen MH, Svane IM. Safety, Immune and Clinical Responses in Metastatic Melanoma Patients Vaccinated With a Long Peptide Derived From Indoleamine 2,3-Dioxygenase in Combination With Ipilimumab. Cytotherapy (2016) 18(8):1043–55. doi: 10.1016/j.jcyt.2016.05.010
    1. Nathan P, Hassel JC, Rutkowski P, Baurain JF, Butler MO, Schlaak M, et al. . Overall Survival Benefit With Tebentafusp in Metastatic Uveal Melanoma. N Engl J Med (2021) 385(13):1196–206. doi: 10.1056/NEJMoa2103485
    1. Atkins MB, Robertson MJ, Gordon M, Lotze MT, DeCoste M, DuBois JS, et al. . Phase I Evaluation of Intravenous Recombinant Human Interleukin 12 in Patients With Advanced Malignancies. Clin Cancer Res (1997) 3(3):409–17.
    1. Portielje JE, Gratama JW, van Ojik HH, Stoter G, Kruit WH. IL-12: A Promising Adjuvant for Cancer Vaccination. Cancer Immunol Immunother (2003) 52(3):133–44. doi: 10.1007/s00262-002-0356-5
    1. Gerer KF, Erdmann M, Hadrup SR, Lyngaa R, Martin LM, Voll RE, et al. . Preclinical Evaluation of NF-kappaB-Triggered Dendritic Cells Expressing the Viral Oncogenic Driver of Merkel Cell Carcinoma for Therapeutic Vaccination. Ther Adv Med Oncol (2017) 9(7):451–64. doi: 10.1177/1758834017712630
    1. Prommersberger S, Hofflin S, Schuler-Thurner B, Schuler G, Schaft N, Dorrie J. A New Method to Monitor Antigen-Specific CD8+ T Cells, Avoiding Additional Target Cells and the Restriction to Human Leukocyte Antigen Haplotype. Gene Ther (2015) 22(6):516–20. doi: 10.1038/gt.2015.15
    1. Nielsen M, Lundegaard C, Blicher T, Lambert K, Harndahl M, Justesen S, et al. . NetMHCpan, a Method for Quantitative Predictions of Peptide Binding to Any HLA-A and -B Locus Protein of Known Sequence. PLoS One (2007) 2(8):e796. doi: 10.1371/journal.pone.0000796
    1. Andersen RS, Kvistborg P, Frosig TM, Pedersen NW, Lyngaa R, Bakker AH, et al. . Parallel Detection of Antigen-Specific T Cell Responses by Combinatorial Encoding of MHC Multimers. Nat Protoc (2012) 7(5):891–902. doi: 10.1038/nprot.2012.037
    1. Danilova L, Anagnostou V, Caushi JX, Sidhom JW, Guo H, Chan HY, et al. . The Mutation-Associated Neoantigen Functional Expansion of Specific T Cells (MANAFEST) Assay: A Sensitive Platform for Monitoring Antitumor Immunity. Cancer Immunol Res (2018) 6(8):888–99. doi: 10.1158/2326-6066.CIR-18-0129

Source: PubMed

3
Sottoscrivi