Peptide Vaccination Against PD-L1 With IO103 a Novel Immune Modulatory Vaccine in Multiple Myeloma: A Phase I First-in-Human Trial

Nicolai Grønne Jørgensen, Uffe Klausen, Jacob Handlos Grauslund, Carsten Helleberg, Thomas Granum Aagaard, Trung Hieu Do, Shamaila Munir Ahmad, Lars Rønn Olsen, Tobias Wirenfeldt Klausen, Marie Fredslund Breinholt, Morten Hansen, Evelina Martinenaite, Özcan Met, Inge Marie Svane, Lene Meldgaard Knudsen, Mads Hald Andersen, Nicolai Grønne Jørgensen, Uffe Klausen, Jacob Handlos Grauslund, Carsten Helleberg, Thomas Granum Aagaard, Trung Hieu Do, Shamaila Munir Ahmad, Lars Rønn Olsen, Tobias Wirenfeldt Klausen, Marie Fredslund Breinholt, Morten Hansen, Evelina Martinenaite, Özcan Met, Inge Marie Svane, Lene Meldgaard Knudsen, Mads Hald Andersen

Abstract

Background: Immune checkpoint blockade with monoclonal antibodies targeting programmed death 1 (PD-1) and its ligand PD-L1 has played a major role in the rise of cancer immune therapy. We have identified naturally occurring self-reactive T cells specific to PD-L1 in both healthy donors and cancer patients. Stimulation with a PD-L1 peptide (IO103), activates these cells to exhibit inflammatory and anti-regulatory functions that include cytotoxicity against PD-L1-expressing target cells. This prompted the initiation of the present first-in-human study of vaccination with IO103, registered at clinicaltrials.org (NCT03042793).

Methods: Ten patients with multiple myeloma who were up to 6 months after high dose chemotherapy with autologous stem cell support, were enrolled. Subcutaneous vaccinations with IO103 with the adjuvant Montanide ISA 51 was given up to fifteen times during 1 year. Safety was assessed by the common toxicity criteria for adverse events (CTCAE). Immunogenicity of the vaccine was evaluated using IFNγ enzyme linked immunospot and intracellular cytokine staining on blood and skin infiltrating lymphocytes from sites of delayed-type hypersensitivity. The clinical course was described.

Results: All adverse reactions to the PD-L1 vaccine were below CTCAE grade 3, and most were grade 1-2 injection site reactions. The total rate of adverse events was as expected for the population. All patients exhibited peptide specific immune responses in peripheral blood mononuclear cells and in skin-infiltrating lymphocytes after a delayed-type hypersensitivity test. The clinical course was as expected for the population. Three of 10 patients had improvements of responses which coincided with the vaccinations.

Conclusion: Vaccination against PD-L1 was associated with low toxicity and high immunogenicity. This study has prompted the initiation of later phase trials to assess the vaccines efficacy.

Clinical trial registration: clinicaltrials.org, identifier NCT03042793.

Keywords: PD-L1; first-in-human; myeloma; peptide; vaccination.

Copyright © 2020 Jørgensen, Klausen, Grauslund, Helleberg, Aagaard, Do, Ahmad, Olsen, Klausen, Breinholt, Hansen, Martinenaite, Met, Svane, Knudsen and Andersen.

Figures

Figure 1
Figure 1
IFNγ-ELISPOT immune responses against IO103. (A) responses in PBMCs in vaccinated patients (bars represent median); (B) Heatmap of responses in PBMCs per vaccinated per time point. (*DFRx1; **DFRx2); (C) representative example of ELISPOT-wells with response; (D) best response in PBMCs in vaccinated patients; (E) responses in unvaccinated reference cohort including time point before HDT (All p-values: Wilcoxon matched-pairs signed rank test); (F) IFNγ-ELISPOT responses against IO103 in bone marrow samples from patient 4.
Figure 2
Figure 2
Immune responses to the vaccine in the skin. (A) IFNγ-ELISPOT responses against the PD-L1 peptide IO103 of skin infiltrating lymphocytes after delayed type hypersensitivity reaction performed after 6 vaccines. 300,000 cells per well. Samples were run in triplicate or quadruplicate. **DFRx2; (B) intracellular cytokine staining (ICS) of SKILs. Almost all cells were CD4+ (not shown). Two out of six evaluable patients had TNFα- and INFγ-double positive SKILs; (C) example of a double positive ICS (patient 7).
Figure 3
Figure 3
Flow cytometric analyses of levels of Tregs. (A) FoxP3+ Treg. (B) CD25highCD127neg Treg; (C) CCR4 + Primed Tregs; (D) CD15s Effector Tregs. % of CD4+ PBMCs. Bars represent median (Mann-Whitney).
Figure 4
Figure 4
Clinical course. Swimmer’s plot. Colors of bars symbolize depth of response at start of vaccinations, after HDT and during the vaccination course. †patient 8 had a rapid relapse after having received 11 vaccinations and died shortly thereafter despite initiation of daratumumab-lenalidomide-dexamethasone.

References

    1. Bedognetti D, Ceccarelli M, Galluzzi L, Lu R, Palucka K, Samayoa J, et al. Toward a comprehensive view of cancer immune responsiveness: A synopsis from the SITC workshop. J Immunother Cancer (2019) 7:1–23. 10.1186/s40425-019-0602-4
    1. Naidoo J, Page DB, Li BT, Connell LC, Schindler K, Lacouture ME, et al. Toxicities of the Anti-PD-1 and Anti-PD-L1 Immune Checkpoint Antibodies. Ann Oncol (2015) 26:2375–91. 10.1093/annonc/mdv383
    1. Munir S, Andersen GH, Met Ö, Donia M, Frøsig TM, Larsen SK, et al. HLA-restricted CTL that are specific for the immune checkpoint ligand PD-L1 occur with high frequency in cancer patients. Cancer Res (2013) 73:1764–76. 10.1158/0008-5472.CAN-12-3507
    1. Munir S, Andersen GH, Woetmann A, Ødum N, Becker JC. Andersen MH.Cutaneous T cell lymphoma cells are targets for immune checkpoint ligand PD-L1-specific, cytotoxic T cells. Leukemia (2013) 27:2251–2253. 10.1038/leu.2013.118
    1. Ahmad SM, Larsen SK, Svane IM, Andersen MH. Harnessing PD-L1-specific cytotoxic T cells for anti-leukemia immunotherapy to defeat mechanisms of immune escape mediated by the PD-1 pathway. Leukemia (2013) 28:236–8. 10.1038/leu.2013.261
    1. Holmström MO, Riley CH, Skov V, Svane IM, Hasselbalch HC. Andersen MH.Spontaneous T-cell responses against the immune check point programmed-death-ligand 1 (PD-L1) in patients with chronic myeloproliferative neoplasms correlate with disease stage and clinical response. Oncoimmunology (2018) 7:1–6. 10.1080/2162402X.2018.1433521
    1. Munir S, Andersen GH, Svane IM, Andersen MH. The immune checkpoint regulator PD-L1 is a specific target for naturally occurring CD4(+) T cells. Oncoimmunology (2013) 2:e23991. 10.4161/onci.23991
    1. Munir Ahmad S, Martinenaite E, Hansen M, Junker N, Borch TH, Met Ö, et al. PD-L1 peptide co-stimulation increases immunogenicity of a dendritic cell-based cancer vaccine. Oncoimmunology (2016) 5:e1202391. 10.1080/2162402X.2016.1202391
    1. Benson DM, Bakan CE, Mishra A, Hofmeister CC, Efebera Y, Becknell B, et al. The PD-1 / PD-L1 axis modulates the natural killer cell versus multiple myeloma effect : a therapeutic target for CT-011, a novel monoclonal anti – PD-1 antibody. Blood (2010) 116:2286–94. 10.1182/blood-2010-02-271874
    1. Liu J, Hamrouni A, Wolowiec D, Coiteux V, Kuliczkowski K, Hetuin D, et al. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-γ and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood (2007) 110:296–304. 10.1182/blood-2006-10-051482
    1. Hallett WHD, Jing W, Drobyski WR, Johnson BD. Immunosuppressive Effects of Multiple Myeloma Are Overcome by PD-L1 Blockade. Biol Blood Marrow Transplant (2011) 17:1133–45. 10.1016/j.bbmt.2011.03.011
    1. Rosenblatt J, Avivi I, Vasir B, Uhl L, Munshi NC, Katz T, et al. Vaccination with dendritic cell/tumor fusions following autologous stem cell transplant induces immunologic and clinical responses in multiple myeloma patients. Clin Cancer Res (2013) 19:3640–8. 10.1158/1078-0432.CCR-13-0282
    1. Ray A, Das DS, Song Y, Richardson P, Munshi NC, Chauhan D, et al. Targeting PD1–PDL1 immune checkpoint in plasmacytoid dendritic cell interactions with T cells, natural killer cells and multiple myeloma cells. Leukemia (2015) 29:1441–4. 10.1038/leu.2015.11
    1. Sponaas AM, Moharrami NN, Feyzi E, Standal T, Rustad EH, Waage A, et al. PDL1 expression on plasma and dendritic cells in myeloma bone marrow suggests benefit of targeted anti PD1-PDL1 therapy. PloS One (2015) 10:e0139867. 10.1371/journal.pone.0139867
    1. An G, Acharya C, Feng X, Wen K, Zhong M, Zhang L, et al. Osteoclasts promote immune suppressive microenvironment in multiple myeloma: therapeutic implication. Blood (2016) 128:1590–1603. 10.1182/blood-2016-03-707547
    1. Paiva B, Azpilikueta A, Puig N, Ocio EM, Sharma R, Oyajobi BO, et al. PD-L1/PD-1 presence in the tumor microenvironment and activity of PD-1 blockade in multiple myeloma. Leukemia (2015) 29:2110–3. 10.1038/leu.2015.79
    1. Ishibashi M, Tamura H, Sunakawa M, Kondo-Onodera A, Okuyama N, Hamada Y, et al. Myeloma Drug Resistance Induced by Binding of Myeloma B7-H1 (PD-L1) to PD-1. Cancer Immunol Res (2016) 4:779–88. 10.1158/2326-6066.CIR-15-0296
    1. Crescenzi A, Annibali O, Bianchi A, Pagano A, Donati M, Grifoni A, et al. PD-1/PD-L1 expression in extra-medullary lesions of multiple myeloma. Leuk Res (2016) 49:98–101. 10.1016/j.leukres.2016.09.008
    1. Gasmi B, Smith E, Dogan A, Hsu M, Devlin S, Pichardo J, et al. Presence of PD-1 Expressing T Cells Predicts for Inferior Overall Survival in Newly Diagnosed Multiple Myeloma. ASH 2015 (abstract 1785) (2015). Available at: (Accessed 10th December 2015).
    1. Tamura H, Ishibashi M, Yamashita T, Tanosaki S, Okuyama N, Kondo A, et al. Marrow stromal cells induce B7-H1 expression on myeloma cells, generating aggressive characteristics in multiple myeloma. Leukemia (2013) 27:464–472. 10.1038/leu.2012.213
    1. Lesokhin AM, Ansell SM, Armand P, Scott EC, Halwani A, Gutierrez M, et al. Nivolumab in Patients with Relapsed or Refractory Lymphoid Malignancy: Preliminary Results of a Phase I Study. J Clin Oncol (2016) 34:2698–704. 10.1200/JCO.2015.65.9789
    1. Usmani SZ, Schjesvold F, Oriol A, Karlin L, Cavo M, Rifkin RM, et al. Pembrolizumab plus lenalidomide and dexamethasone for patients with treatment-naive multiple myeloma (KEYNOTE-185): a randomised, open-label, phase 3 trial. Lancet Haematol (2019) 6:e448–58. 10.1016/s2352-3026(19)30109-7
    1. Mateos M-V, Blacklock H, Schjesvold F, Oriol A, Simpson D, George A, et al. Pembrolizumab plus pomalidomide and dexamethasone for patients with relapsed or refractory multiple myeloma (KEYNOTE-183): a randomised, open-label, phase 3 trial. Lancet Haematol (2019) 6:e459–69. 10.1016/s2352-3026(19)30110-3
    1. van der Burg SH, Arens R, Ossendorp F, van Hall T, Melief CJM. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat Rev Cancer (2016) 16:219–233. 10.1038/nrc.2016.16
    1. Chung DJ, Pronschinske KB, Shyer JA, Sharma S, Leung S, Curran SA, et al. T-cell Exhaustion in Multiple Myeloma Relapse after Autotransplant: Optimal Timing of Immunotherapy. Cancer Immunol Res (2016) 4:61–71. 10.1158/2326-6066.CIR-15-0055
    1. Svane IM, Nikolajsen K, Johnsen HE. Antigen-specific T-cell immunity in multiple myeloma patients is restored following high-dose therapy: implications for timing of vaccination. Scand J Immunol (2007) 66:465–75. 10.1111/j.1365-3083.2007.01993.x
    1. Williams KM, Hakim FT, Gress RE. T cell immune reconstitution following lymphodepletion. Semin Immunol (2007) 19:318–330. 10.1016/j.smim.2007.10.004
    1. Gökbuget N, Canaani J, Nagler A, Bishop M, Kröger N. Avigan D.Prevention and treatment of relapse after stem cell transplantation with immunotherapy. Bone Marrow Transplant (2018) 53:664–72. 10.1038/s41409-018-0232-3
    1. Melief CJM, van der Burg SH. Immunotherapy of established (pre)malignant disease by synthetic long peptide vaccines. Nat Rev Cancer (2008) 8:351–60. 10.1038/nrc2373
    1. Ascarateil S, Puget A, Koziol M-E. Safety data of Montanide ISA 51 VG and Montanide ISA 720 VG, two adjuvants dedicated to human therapeutic vaccines. J Immunother Cancer (2015) 3:2015. 10.1186/2051-1426-3-S2-P428
    1. Rajkumar SV, Harousseau JL, Durie B, Andersen KC, Dimopoulos M, Kyle R, et al. Consensus recommendations for the uniform reporting of clinical trials: report of the International Myeloma Workshop Consensus Panel 1. Blood (2011) 117:4691–5. 10.1182/blood-2010-10-299487
    1. de Vries IJM, Bernsen MR, Lesterhuis WJ, Scharenborg NM, Strijk SP, Gerritsen MJP, et al. Immunomonitoring tumor-specific T cells in delayed-type hypersensitivity skin biopsies after dendritic cell vaccination correlates with clinical outcome. J Clin Oncol (2005) 23:5779–87. 10.1200/JCO.2005.06.478
    1. McCutcheon M, Wehner N, Wensky A, Kushner M, Doan S, Hsiao L, et al. A sensitive ELISPOT assay to detect low-frequency human T lymphocytes. J Immunol Methods (1997) 210:149–66. 10.1016/S0022-1759(97)00182-8
    1. Moodie Z, Price L, Janetzki S, Britten CM. Response Determination Criteria for ELISPOT: Toward a Standard that Can Be Applied Across Laboratories. Methods Mol Biol (2012) 792:185–96. 10.1007/978-1-61779-325-7_15
    1. Yang B, Parsha K, Schaar K, Satani N, Xi X, Aronowski J, et al. Cryopreservation of Bone Marrow Mononuclear Cells Alters Their Viability and Subpopulation Composition but Not Their Treatment Effects in a Rodent Stroke Model. Stem Cells Int (2016) 2016:5876836. 10.1155/2016/5876836
    1. Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep (2017) 19:1189–201. 10.1016/j.celrep.2017.04.031
    1. Munir S, Lundsager MT, Jørgensen MA, Hansen M, Petersen TH, Bonefeld CM, et al. Inflammation induced PD-L1-specific T cells. Cell Stress (2019) 3:319–327. 10.15698/cst2019.10.201
    1. Iversen TZ, Engell-Noerregaard L, Ellebaek E, Andersen R, Larsen SK, Bjoern J, et al. Long-lasting disease stabilization in the absence of toxicity in metastatic lung cancer patients vaccinated with an epitope derived from indoleamine 2,3 dioxygenase. Clin Cancer Res (2014) 20:221–32. 10.1158/1078-0432.CCR-13-1560
    1. Sørensen RB, Hadrup SR, Svane IM, Hjortsø MC, Straten PT. Andersen MH.Indoleamine 2,3-dioxygenase specific, cytotoxic T cells as immune regulators. Blood (2011) 117:2200–10. 10.1182/blood-2010-06-288498
    1. Andersen MH. The balance players of the adaptive immune system. Cancer Res (2018) 78:1379–1382. 10.1158/0008-5472.CAN-17-3607
    1. Danish Myeloma Study Group Danish Myeloma Database. (2017). Available at .
    1. Gonsalves WI, Gertz MA, Dispenzieri A, Lacy MQ, Lin Y, Singh PP, et al. Implications of continued response after autologous stem cell transplantation for multiple myeloma. Blood (2013) 122:1746–49. 10.1182/blood-2013-03-492678
    1. Fernández De Larrea C, Dávila J, Isola I, Ocio EM, Rosinol L, García-Sanz R, et al. Absence of spontaneous response improvement beyond day +100 after autologous stem cell transplantation in multiple myeloma. Bone Marrow Transplant (2017) 52:567–9. 10.1038/bmt.2016.299
    1. Tran T, Blanc C, Granier C, Saldmann A, Tanchot C. Tartour E.Therapeutic cancer vaccine: building the future from lessons of the past. Semin Immunopathol (2018) 41:69–85. 10.1007/s00281-018-0691-z
    1. van der Burg SH. Correlates of immune and clinical activity of novel cancer vaccines. Semin Immunol (2018) 39:119–36. 10.1016/j.smim.2018.04.001

Source: PubMed

Подписаться