Arginase-1 targeting peptide vaccine in patients with metastatic solid tumors - A phase I trial

Cathrine Lund Lorentzen, Evelina Martinenaite, Julie Westerlin Kjeldsen, Rikke Boedker Holmstroem, Sofie Kirial Mørk, Ayako Wakatsuki Pedersen, Eva Ehrnrooth, Mads Hald Andersen, Inge Marie Svane, Cathrine Lund Lorentzen, Evelina Martinenaite, Julie Westerlin Kjeldsen, Rikke Boedker Holmstroem, Sofie Kirial Mørk, Ayako Wakatsuki Pedersen, Eva Ehrnrooth, Mads Hald Andersen, Inge Marie Svane

Abstract

Background: Arginase-1-producing cells inhibit T cell-mediated anti-tumor responses by reducing L-arginine levels in the tumor microenvironment. T cell-facilitated elimination of arginase-1-expressing cells could potentially restore L-arginine levels and improve anti-tumor responses. The activation of arginase-1-specific T cells may convert the immunosuppressive tumor microenvironment and induce or strengthen local Th1 inflammation. In the current clinical study, we examined the safety and immunogenicity of arginase-1-based peptide vaccination.

Methods: In this clinical phase I trial, ten patients with treatment-refractory progressive solid tumors were treated. The patients received an arginase-1 peptide vaccine comprising three 20-mer peptides from the ARG1 immunological "hot spot" region in combination with the adjuvant Montanide ISA-51. The vaccines were administered subcutaneously every third week (maximum 16 vaccines). The primary endpoint was to evaluate safety assessed by Common Terminology Criteria for Adverse Events 4.0 and laboratory monitoring. Vaccine-specific immune responses were evaluated using enzyme-linked immune absorbent spot assays and intracellular cytokine staining on peripheral blood mononuclear cells. Clinical responses were evaluated using Response Evaluation Criteria in Solid Tumors 1.1.

Results: The vaccination was feasible, and no vaccine-related grade 3-4 adverse events were registered. Nine (90%) of ten patients exhibited peptide-specific immune responses in peripheral blood mononuclear cells. Six (86%) of the seven evaluable patients developed a reactive T cell response against at least one of the ARG1 peptides during treatment. A phenotypic classification revealed that arginase-1 vaccine-specific T cells were both CD4+ T cells and CD8+ T cells. Two (20%) of ten patients obtained stable disease for respectively four- and seven months on vaccination treatment.

Conclusion: The peptide vaccine against arginase-1 was safe. Nine (90%) of ten patients had measurable peptide-specific responses in the periphery blood, and two (20%) of ten patients attained stable disease on protocol treatment.

Clinical trial registration: https://ichgcp.net/clinical-trials-registry/NCT03689192, identifier NCT03689192.

Keywords: arginase-1; first-in-human; peptide; solid tumors; vaccination.

Conflict of interest statement

MA has various patent applications in relation to the therapeutic uses of ARG1 peptides. The patents are allocated to the company IO Biotech. MA is a founder, advisor, and shareholder for IO Biotech. EM, AP, and EE are employees at IO Biotech. IS has lectured for or had advisory board relationships with MSD, Sanofi Aventis, BMS, Pierre Fabre, Novartis, TILT Biotherapeutics, IO Biotech, and Novo Nordisk. IS has received research grants from Lytix biopharma, IO Biotech, BMS, Adaptimmune, and TILT Biotherapeutics. IS is a co-founder and shareholder for the company IO Biotech. 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 Lorentzen, Martinenaite, Kjeldsen, Holmstroem, Mørk, Pedersen, Ehrnrooth, Andersen and Svane.

Figures

Figure 1
Figure 1
Consort diagram. Twenty patients with metastatic solid tumors were assessed for eligibility. Thirteen patients were enrolled and received the study treatment. Three patients received

Figure 2

Heatmaps of detected specific arginase-1…

Figure 2

Heatmaps of detected specific arginase-1 (ARG1) responses in peripheral blood mononuclear cells (PBMCs)…

Figure 2
Heatmaps of detected specific arginase-1 (ARG1) responses in peripheral blood mononuclear cells (PBMCs) at baseline and on treatment as measured by interferon (IFN)-γ enzyme-linked immunospot (ELISPOT) assay (n=10). Background has been subtracted. *Indicates positive responses based on Distribution-free Resampling (DFR) method.

Figure 3

CD4+ and CD8+ arginase-1 (ARG1)…

Figure 3

CD4+ and CD8+ arginase-1 (ARG1) vaccine-specific T cell responses in blood. Total ARG1-specific…

Figure 3
CD4+ and CD8+ arginase-1 (ARG1) vaccine-specific T cell responses in blood. Total ARG1-specific CD4+ (black) and CD8+ (grey) T cell responses in peripheral blood mononuclear cells (PBMCs) at baseline and on treatment. The data were quantified by flow cytometry by an increased expression of interferon (IFN)γ, IFNγ + TNFα, and TNFα after five-hour peptide stimulation. A detailed cytokine expression profile is shown in Supplementary Figure 1. The values indicate specific responses subsequent to substraction of background values (n=10). N/A, Not available.

Figure 4

Spider plot showing changes in…

Figure 4

Spider plot showing changes in tumor size on evaluation scans for each patient.…

Figure 4
Spider plot showing changes in tumor size on evaluation scans for each patient. Patients were evaluated every three months. Numbers indicate patient ID. SD, stable disease; PD, progressive disease.

Figure 5

Overall survival and progression-free survival.…

Figure 5

Overall survival and progression-free survival. mPFS, median progression-free survival; mOS, median overall survival.

Figure 5
Overall survival and progression-free survival. mPFS, median progression-free survival; mOS, median overall survival.
Figure 2
Figure 2
Heatmaps of detected specific arginase-1 (ARG1) responses in peripheral blood mononuclear cells (PBMCs) at baseline and on treatment as measured by interferon (IFN)-γ enzyme-linked immunospot (ELISPOT) assay (n=10). Background has been subtracted. *Indicates positive responses based on Distribution-free Resampling (DFR) method.
Figure 3
Figure 3
CD4+ and CD8+ arginase-1 (ARG1) vaccine-specific T cell responses in blood. Total ARG1-specific CD4+ (black) and CD8+ (grey) T cell responses in peripheral blood mononuclear cells (PBMCs) at baseline and on treatment. The data were quantified by flow cytometry by an increased expression of interferon (IFN)γ, IFNγ + TNFα, and TNFα after five-hour peptide stimulation. A detailed cytokine expression profile is shown in Supplementary Figure 1. The values indicate specific responses subsequent to substraction of background values (n=10). N/A, Not available.
Figure 4
Figure 4
Spider plot showing changes in tumor size on evaluation scans for each patient. Patients were evaluated every three months. Numbers indicate patient ID. SD, stable disease; PD, progressive disease.
Figure 5
Figure 5
Overall survival and progression-free survival. mPFS, median progression-free survival; mOS, median overall survival.

References

    1. Maeda H, Khatami M. Analyses of repeated failures in cancer therapy for solid tumors: poor tumor-selective drug delivery, low therapeutic efficacy and unsustainable costs. Clin Transl Med (2018) 7:11. doi: 10.1186/s40169-018-0185-6
    1. Nixon NA, Blais N, Ernst S, Kollmannsberger C, Bebb G, Butler M, et al. . Current landscape of immunotherapy in the treatment of solid tumours, with future opportunities and challenges. Curr Oncol (2018) 25:e373–84. doi: 10.3747/co.25.3840
    1. Lindau D, Gielen P, Kroesen M, Wesseling P, Adema GJ. The immunosuppressive tumour network: Myeloid-derived suppressor cells, regulatory T cells and natural killer T cells. Immunology (2013) 138:105–15. doi: 10.1111/imm.12036
    1. Geiger R, Rieckmann JC, Wolf T, Basso C, Feng Y, Fuhrer T, et al. . L-arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell (2016) 167:829–842.e13. doi: 10.1016/j.cell.2016.09.031
    1. Kim PS, Iyer RK, Lu KV, Yu H, Karimi A, Kern RM, et al. . Expression of the liver form of arginase in erythrocytes. Mol Genet Metab (2002) 76:100–10. doi: 10.1016/S1096-7192(02)00034-3
    1. Rodriguez PC, Quiceno DG, Ochoa AC. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood (2007) 109:1568–73. doi: 10.1182/blood-2006-06-031856
    1. Rodriguez PC, Zea AH, DeSalvo J, Culotta KS, Zabaleta J, Quiceno DG, et al. . L-arginine consumption by macrophages modulates the expression of CD3ζ chain in T lymphocytes. J Immunol (2003) 171:1232–9. doi: 10.4049/jimmunol.171.3.1232
    1. Coosemans A, Decoene J, Baert T, Laenen A, Kasran A, Verschuere T, et al. . Immunosuppressive parameters in serum of ovarian cancer patients change during the disease course. Oncoimmunology (2016) 5:e1111505. doi: 10.1080/2162402X.2015.1111505
    1. Rodriguez PC, Ernstoff MS, Hernandez C, Atkins M, Zabaleta J, Sierra R, et al. . Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res (2009) 69:1553–60. doi: 10.1158/0008-5472.CAN-08-1921
    1. Singh R, Pervin S, Karimi A, Cederbaum S, Chaudhuri G. Arginase activity in human breast cancer cell lines: N(omega)-hydroxy-L-arginine selectively inhibits cell proliferation and induces apoptosis in MDA-MB-468 cells. Cancer Res (2000) 60:3305–12. doi: 10.4161/onci.21678
    1. Lang S, Bruderek K, Kaspar C, Höing B, Kanaan O, Dominas N, et al. . Clinical relevance and suppressive capacity of human myeloid-derived suppressor cell subsets. Clin Cancer Res (2018) 24:4834–44. doi: 10.1158/1078-0432.CCR-17-3726
    1. Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. . TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature (2018) 554:544–8. doi: 10.1038/nature25501
    1. Martinenaite E, Mortensen REJ, Hansen M, Orebo Holmström M, Munir Ahmad S, Grønne Dahlager Jørgensen N, et al. . Frequent adaptive immune responses against arginase-1. Oncoimmunology (2018) 7:e1404215. doi: 10.1080/2162402X.2017.1404215
    1. Martinenaite E, Ahmad SM, Svane IM, Andersen MH. Peripheral memory T cells specific for arginase-1. Cell Mol Immunol (2019) 16:718–9. doi: 10.1038/s41423-019-0231-3
    1. Andersen MH. The balance players of the adaptive immune system. Cancer Res (2018) 78:1379–82. doi: 10.1158/0008-5472.CAN-17-3607
    1. Martinenaite E, Ahmad SM, Bendtsen SK, Jørgensen MA, Weis-Banke SE, Svane IM, et al. . Arginase-1-based vaccination against the tumor microenvironment: the identification of an optimal T-cell epitope. Cancer Immunol Immunother (2019) 68:1901–7. doi: 10.1007/s00262-019-02425-6
    1. Kjeldsen JW, Lorentzen CL, Martinenaite E, Ellebaek E, Donia M, Holmstroem RB, et al. . A phase 1/2 trial of an immune-modulatory vaccine against IDO/PD-L1 in combination with nivolumab in metastatic melanoma. Nat Med (2021) 27:2212–23. doi: 10.1038/s41591-021-01544-x
    1. Jørgensen NG, Klausen U, Grauslund JH, Helleberg C, Aagaard TG, Do TH, et al. . Peptide vaccination against PD-L1 with IO103 a novel immune modulatory vaccine in multiple myeloma: A phase I first-in-Human trial. Front Immunol (2020) 11:595035. doi: 10.3389/fimmu.2020.595035
    1. Handlos Grauslund J, Holmström MO, Jørgensen NG, Klausen U, Weis-Banke SE, El Fassi D, et al. . Therapeutic cancer vaccination with a peptide derived from the calreticulin exon 9 mutations induces strong cellular immune responses in patients with CALR-mutant chronic myeloproliferative neoplasms. Front Oncol (2021) 11:637420. doi: 10.3389/fonc.2021.637420
    1. Steggerda SM, Bennett MK, Chen J, Emberley E, Huang T, Janes JR, et al. . Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment. J Immunother Cancer (2017) 5:101. doi: 10.1186/s40425-017-0308-4
    1. Montero AJ, Diaz-Montero CM, Kyriakopoulos CE, Bronte V, Mandruzzato S. Myeloid-derived suppressor cells in cancer patients: a clinical perspective. J Immunother (2012) 35:107–15. doi: 10.1097/CJI.0b013e318242169f
    1. Solito S, Marigo I, Pinton L, Damuzzo V, Mandruzzato S, Bronte V. Myeloid-derived suppressor cell heterogeneity in human cancers. Ann N Y Acad Sci (2014) 1319:47–65. doi: 10.1111/nyas.12469
    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. doi: 10.1007/978-1-61779-325-7_15
    1. Naing A, Bauer T, Papadopoulos KP, Rahma O, Tsai F, Garralda E, et al. . Phase I study of the arginase inhibitor INCB001158 (1158) alone and in combination with pembrolizumab (PEM) in patients (Pts) with advanced/metastatic (adv/met) solid tumours. Ann Oncol (2019) 30:v160. doi: 10.1093/annonc/mdz244.002
    1. Saxena M, van der Burg SH, Melief CJM, Bhardwaj N. Therapeutic cancer vaccines. Nat Rev Cancer (2021) 21:360–78. doi: 10.1038/s41568-021-00346-0
    1. Ugel S, De Sanctis F, Mandruzzato S, Bronte V. Tumor-induced myeloid deviation: When myeloid-derived suppressor cells meet tumor-associated macrophages. J Clin Invest (2015) 125:3365–76. doi: 10.1172/JCI80006
    1. Jørgensen MA, Ugel S, Hübbe ML, Carretta M, Perez-Penco M, Weis-Banke SE, et al. . Arginase 1–based immune modulatory vaccines induce anticancer immunity and synergize with anti–PD-1 checkpoint blockade. Cancer Immunol Res (2021) 9:1316–26. doi: 10.1158/2326-6066.CIR-21-0280

Source: PubMed

3
Předplatit